1
|
Dávila Saldaña BJ, Schultz KR, Ramgopal A, Boiko JR, Beebe K, Carpenter PA, Chan SS, Paczesny S, Aguayo-Hiraldo P, Cuvelier GDE, Rotz SJ, Duncan CN, Williams KM. Pediatric Transplant and Cellular Therapy Consortium RESILIENT Conference on Pediatric Chronic Graft-versus-Host Disease Survivorship after Hematopoietic Cell Transplantation: Part II. Organ Dysfunction and Immune Reconstitution Considerations for Children with Chronic Graft-versus-Host Disease after Hematopoietic Cell Transplantation. Transplant Cell Ther 2025:S2666-6367(25)00913-3. [PMID: 39855565 DOI: 10.1016/j.jtct.2025.01.885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
While highly morbid forms of chronic graft versus host disease (cGVHD) and severe late effects of allogeneic hematopoietic cell transplantation (HCT) can impact children and adults alike, unique considerations arise in pediatric cases regarding diagnosis, monitoring, treatment, and likelihood of resolution. As children can present with atypical features of cGVHD and with more significant disease due to inability to communicate symptoms, they may be at increased risk for highly morbid forms of cGVHD and incur greater subsequent late effects, which may be more pronounced in those with underlying chromosomal breakage syndromes, with higher prevalence in pediatric HCT recipients. The long-term effects of cGVHD and its therapies include impaired immune reconstitution, leading to increased risks of infection and secondary malignant neoplasms. However, children also have the greatest potential for full immune reconstitution, due to thymus recovery that could impact the timing of vaccination with respect to tolerance and restoration of optimal immunity. Developing strategies to mitigate the late effects incurred with, and as a result of, cGVHD is of critical importance. The working group recommends surveillance strategies for late effects in patients with cGVHD, increased utilization of emerging diagnostic tools, integration of monitoring for cGVHD treatment response, and development of new treatments and specifies aims of future research endeavors.
Collapse
Affiliation(s)
- Blachy J Dávila Saldaña
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, District of Columbia.
| | - Kirk R Schultz
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Archana Ramgopal
- Department of Pediatric Bone Marrow Transplantation & Cellular Therapy, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Julie R Boiko
- Translational Science and Therapeutics Division, Fred Hutch Cancer Center, Seattle, Washington
| | - Kristen Beebe
- Blood and Marrow Transplantation, Mayo Clinic Arizona and Phoenix Children's Hospital, Phoenix, Arizona
| | - Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Sherwin S Chan
- Department of Radiology, Children's Mercy Kansas City, Kansas City, Missouri
| | - Sophie Paczesny
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Paibel Aguayo-Hiraldo
- Cancer and Blood Disorders Institute, Children's Hospital Los Angeles, University Southern California Keck School of Medicine, Los Angeles, California
| | - Geoffrey D E Cuvelier
- Department of Pediatric Oncology and Transplantation, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Seth J Rotz
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, Ohio
| | - Christine N Duncan
- Department of Pediatric Hematology-Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kirsten M Williams
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| |
Collapse
|
2
|
Pashkina E, Blinova E, Bykova M, Aktanova A, Denisova V. Cell Therapy as a Way to Increase the Effectiveness of Hematopoietic Stem Cell Transplantation. Cells 2024; 13:2056. [PMID: 39768148 PMCID: PMC11675046 DOI: 10.3390/cells13242056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a standard method for treating a number of pathologies, primarily blood diseases. Timely restoration of the immune system after HSCT is a critical factor associated with the development of complications such as relapses or secondary tumors and various infections, as well as the graft-versus-host reaction in allogeneic transplantation, which ultimately affects the survival of patients. Introduction into the recipient's body of immune system cells that are incapable of sensitization by recipient antigens during the period of immune reconstitution can increase the rate of restoration of the immune system, as well as reduce the risk of complications. This review presents the results of studies on cell therapy with various cell subpopulations of both bone marrow and mesenchymal origin during HSCT.
Collapse
Affiliation(s)
- Ekaterina Pashkina
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
- Department of Clinical Immunology, Novosibirsk State Medical University, 52, Krasny Prospect, 630091 Novosibirsk, Russia
| | - Elena Blinova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
| | - Maria Bykova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
| | - Alina Aktanova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
- Department of Clinical Immunology, Novosibirsk State Medical University, 52, Krasny Prospect, 630091 Novosibirsk, Russia
| | - Vera Denisova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
| |
Collapse
|
3
|
Zhang J, Guan X, Zhong X. Immunosenescence in digestive system cancers: Mechanisms, research advances, and therapeutic strategies. Semin Cancer Biol 2024; 106-107:234-250. [PMID: 39510149 DOI: 10.1016/j.semcancer.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/21/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
Increasing lifespans and external environmental factors have contributed to the increase of age-related diseases, particularly cancer. A decrease in immune surveillance and clearance of cancer cells is the result of immunosenescence, which involves the remodeling of immune organs, the changes and functional decline of immune cell subsets, in association with systemic low-grade chronic inflammation. Stem cells aging in bone marrow and thymic involution are the most important causes of immunosenescence. Senescent cancer cells promote the differentiation, recruitment, and functional upregulation of immune-suppressive cell subsets e.g. regulatory T cells (Tregs), myeloid-derived suppressor cell (MDSC), tumor-associated macrophages (TAMS) through senescence-associated secretory phenotype (SASP) further exacerbating the immunosuppressive microenvironment. For digestive system cancers, age-related damage to the intestinal mucosal barrier, the aging of gut-associated lymphoid tissue (GALT), exposure to xenobiotic stimuli throughout life, and dysbiosis make the local immune microenvironment more vulnerable. This article systematically reviews the research progress of immunosenescence and immune microenvironment in digestive system cancers, as well as the exploration of related therapy strategies, hoping to point out new directions for research in the digestive system cancers.
Collapse
Affiliation(s)
- Junyan Zhang
- Department of Surgical Oncology and General Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaojiao Guan
- Department of Pathology, Shengjing Hospital, China Medical University, Shenyang, China.
| | - Xinwen Zhong
- Department of Thoracic Surgery, First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Justus JLP, Beltrame MP, de Azambuja AP, Schluga YC, Martins EA, Rocha MTL, Rodrigues AM, Loth G, Lima ACM, Bonfim C. Immune recovery and the role of recent thymic emigrated T lymphocytes after pediatric hematopoietic stem cell transplantation. Cytotherapy 2024; 26:980-987. [PMID: 38762804 DOI: 10.1016/j.jcyt.2024.04.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND AIMS Adequate re-establishment of thymopoiesis is critical for long-term immune reconstitution after hematopoietic cell transplantation (HCT), potentially impacting patient survival rates. This study aimed to evaluate immune reconstitution in pediatric HCT recipients by quantifying recent thymic emigrants (RTEs), specifically CD3+CD31+CD45RA+ cells. METHODS We conducted a retrospective analysis of 186 pediatric patients transplanted between 2013 and 2020, undergoing their first allogeneic HCT, who were alive in the first 100 days after transplantation with immune recovery evaluation at three time points: day 100, day 180 and day 360 after HCT. We analyzed the distribution of peripheral blood subsets of T, B and natural killer lymphocytes and assessed the impact of underlying disease, HCT type, stem cell source, recipient age, conditioning regimen, graft-versus-host disease (GVHD) occurrence and cytomegalovirus (CMV) reactivation on immune recovery. RESULTS At day 100, patients under 10 years exhibited higher RTE CD4+ and CD8+CD31+CD45RA+ counts compared with older patients (5.3 versus 2.2 cells/µL, P = 0.022 and 48 versus 72.8 cells/µL, P = 0.049, respectively). Patients with haploidentical HCT had lower RTE CD4+ counts compared with those with unrelated or related donors (2.4 versus 4.4 versus 7.9 cells/µL, P = 0.024). Administration of rabbit anti-thymocyte globulin negatively impacted RTE CD4+ production (median, 6.5 versus 2.4 cells/µL, P = 0.007). At day 180, the presence of GVHD had a negative influence on RTE production (11.7 versus 56.8 cells/µL, P < 0.001), particularly higher-grade acute GVHD (without, 56.8 cells/µL, grade 1-2, 28.1 cells/µL, grade 3-4, 6.0 cells/µL, P < 0.001). Patients with CMV reactivation had higher CD8+CD31+CD45RA+ compared with those without reactivation (median, 204.6 versus 100.2 cells/µL, P = 0.022). At day 360, no variables significantly affected RTE recovery. Overall survival at 5-year follow-up was 87.7%, with a median of 1170 days (range, 122-3316). Multivariate analysis showed that age >10 years (P = 0.038), negative CMV donor serology (P = 0.0029) and acute GVHD (P = 0.0026) had a negative impact on survival. CONCLUSIONS This study highlights variations in RTE production based on patient age, donor type and immunosuppression regimen employed.
Collapse
Affiliation(s)
- Julie Lillian Pimentel Justus
- Flow Cytometry Laboratory, Clinics Hospital, Federal University of Paraná, Curitiba, Brazil; Post-Graduation Program in Children and Adolescent Health, Clinics Hospital, Federal University of Parana, Curitiba, Brazil.
| | - Miriam P Beltrame
- Flow Cytometry Laboratory, Clinics Hospital, Federal University of Paraná, Curitiba, Brazil; Post-Graduation Program in Children and Adolescent Health, Clinics Hospital, Federal University of Parana, Curitiba, Brazil
| | - Ana Paula de Azambuja
- Flow Cytometry Laboratory, Clinics Hospital, Federal University of Paraná, Curitiba, Brazil
| | - Yara C Schluga
- Flow Cytometry Laboratory, Clinics Hospital, Federal University of Paraná, Curitiba, Brazil
| | - Edna A Martins
- Flow Cytometry Laboratory, Clinics Hospital, Federal University of Paraná, Curitiba, Brazil
| | | | - Adriana Mello Rodrigues
- Pediatric Blood and Marrow Transplantation Division, Clinics Hospital, Federal University of Paraná, Curitiba, Brazil
| | - Gisele Loth
- Pediatric Blood and Marrow Transplantation Division, Clinics Hospital, Federal University of Paraná, Curitiba, Brazil
| | | | - Carmem Bonfim
- Post-Graduation Program in Children and Adolescent Health, Clinics Hospital, Federal University of Parana, Curitiba, Brazil; Pediatric Blood and Marrow Transplantation Division, Hospital Pequeno Príncipe, Curitiba, Brazil
| |
Collapse
|
5
|
Kielsen K, Møller DL, Pedersen AE, Nielsen CH, Ifversen M, Ryder LP, Müller K. Cytomegalovirus infection is associated with thymic dysfunction and chronic graft-versus-host disease after pediatric hematopoietic stem cell transplantation. Clin Immunol 2024; 265:110302. [PMID: 38942161 DOI: 10.1016/j.clim.2024.110302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Pediatric hematopoietic stem cell transplantation (HSCT) is challenged by chronic graft-versus-host disease (cGvHD) significantly affecting survival and long-term morbidity, but underlying mechanisms including the impact of post-HSCT CMV infection are sparsely studied. We first investigated the impact of CMV infection for development of cGvHD in 322 children undergoing standard myeloablative HSCT between 2000 and 2018. Clinically significant CMV infection (n = 61) was an independent risk factor for chronic GvHD in a multivariable Cox regression analysis (HR = 2.17, 95% CI = 1.18-3.97, P = 0.013). We next explored the underlying mechanisms in a subcohort of 39 children. CMV infection was followed by reduced concentration of recent thymic emigrants (17.5 vs. 51.9 × 106/L, P = 0.048) and naïve CD4+ and CD8+ T cells at 6 months post-HSCT (all P < 0.05). Furthermore, CD25highFOXP3+ Tregs tended to be lower in patients with CMV infection (2.9 vs. 9.6 × 106/L, P = 0.055), including Tregs expressing the naivety markers CD45RA and Helios. CD8+ T-cell numbers rose after CMV infection and was dominated by exhausted PD1-expressing cells (66% vs. 39%, P = 0.023). These findings indicate that post-HSCT CMV infection is a main risk factor for development of chronic GvHD after pediatric HSCT and suggest that this effect is caused by reduced thymic function with a persistently impaired production of naïve and regulatory T cells in combination with increased peripheral T-cell exhaustion.
Collapse
Affiliation(s)
- Katrine Kielsen
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Departmen of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.
| | - Dina Leth Møller
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Claus Henrik Nielsen
- Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Odontology, University of Copenhagen, Copenhagen, Denmark
| | - Marianne Ifversen
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Lars Peter Ryder
- Departmen of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Klaus Müller
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Institute for Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Uhlemann H, Epp K, Klesse C, Link-Rachner CS, Surendranath V, Günther UP, Schetelig J, Heidenreich F. Shape of the art: TCR-repertoire after allogeneic hematopoietic cell transplantation. Best Pract Res Clin Haematol 2024; 37:101558. [PMID: 39098804 DOI: 10.1016/j.beha.2024.101558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/03/2024] [Accepted: 06/27/2024] [Indexed: 08/06/2024]
Abstract
The human adaptive immune repertoire is characterized by specificity and diversity to provide immunity against past and future tasks. Such tasks are mainly infections but also malignant transformations of cells. With its multiple lines of defense, the human immune system contains both, rapid reaction forces and the potential to capture, disassemble and analyze strange structures in order to teach the adaptive immune system and mount a specific immune response. Prevention and mitigation of autoimmunity is of equal importance. In the context of allogeneic hematopoietic cell transplantation (HCT) specific challenges exist with the transfer of cells from the adapted donor immune system to the immunosuppressed recipient. Those challenges are immunogenetic disparity between donor and host, reconstitution of immunity early after HCT by expansion of mature immune effector cells, and impaired thymic function, if the recipient is an adult (as it is the case in most HCTs). The possibility to characterize the adaptive immune repertoire by massively parallel sequencing of T-cell receptor gene rearrangements allows for a much more detailed characterization of the T-cell repertoire. In addition, high-dimensional characterization of immune effector cells based on their immunophenotype and single cell RNA sequencing allow for much deeper insights in adaptive immune responses. We here review, existing - still incomplete - information on immune reconstitution after allogeneic HCT. Building on the technological advances much deeper insights into immune recovery after HCT and adaptive immune responses and can be expected in the coming years.
Collapse
Affiliation(s)
- Heike Uhlemann
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany.
| | - Katharina Epp
- University Hospital Carl Gustav Carus, Dresden, Germany
| | | | | | | | | | - Johannes Schetelig
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany
| | - Falk Heidenreich
- University Hospital Carl Gustav Carus, Dresden, Germany; DKMS Group gGmbH, Clinical Trials Unit, Dresden, Germany
| |
Collapse
|
7
|
Zhou G, Zhan Q, Huang L, Dou X, Cui J, Xiang L, Qi Y, Wu S, Liu L, Xiao Q, Chen J, Tang X, Zhang H, Wang X, Luo X, Ren G, Yang Z, Liu L, Yan X, Luo Q, Pei C, Dai Y, Zhu Y, Zhou H, Ren G, Wang L. The dynamics of B-cell reconstitution post allogeneic hematopoietic stem cell transplantation: A real-world study. J Intern Med 2024; 295:634-650. [PMID: 38439117 DOI: 10.1111/joim.13776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
BACKGROUND The immune reconstitution after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is crucial for preventing infections and relapse and enhancing graft-versus-tumor effects. B cells play an important role in humoral immunity and immune regulation, but their reconstitution after allo-HSCT has not been well studied. METHODS In this study, we analyzed the dynamics of B cells in 252 patients who underwent allo-HSCT for 2 years and assessed the impact of factors on B-cell reconstitution and their correlations with survival outcomes, as well as the development stages of B cells in the bone marrow and the subsets in the peripheral blood. RESULTS We found that the B-cell reconstitution in the bone marrow was consistent with the peripheral blood (p = 0.232). B-cell reconstitution was delayed by the male gender, age >50, older donor age, the occurrence of chronic and acute graft-versus-host disease, and the infections of fungi and cytomegalovirus. The survival analysis revealed that patients with lower B cells had higher risks of death and relapse. More importantly, we used propensity score matching to obtain the conclusion that post-1-year B-cell reconstitution is better in females. Meanwhile, using mediation analysis, we proposed the age-B cells-survival axis and found that B-cell reconstitution at month 12 posttransplant mediated the effect of age on patient survival (p = 0.013). We also found that younger patients showed more immature B cells in the bone marrow after transplantation (p = 0.037). CONCLUSION Our findings provide valuable insights for optimizing the management of B-cell reconstitution and improving the efficacy and safety of allo-HSCT.
Collapse
Affiliation(s)
- Guangyu Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Qian Zhan
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Lingle Huang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Xi Dou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Jin Cui
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Lin Xiang
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Yuhong Qi
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Sicen Wu
- Health Management Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Qing Xiao
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Jianbin Chen
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Xiaoqiong Tang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Hongbin Zhang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Xin Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Xiaohua Luo
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Zesong Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Lanxiang Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Xinyu Yan
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Qin Luo
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Caixia Pei
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Yulian Dai
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Yu Zhu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Hao Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Guilin Ren
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
8
|
Major-Monfried H, Hosszu K, McAvoy DP, Vallone A, Shukla N, Gillio A, Spitzer B, Kung AL, Cancio M, Curran K, Scaradavou A, Oved JH, O'Reilly RJ, Boelens JJ, Harris AC. Two novel assays demonstrate persistent daratumumab exposure in a pediatric patient with delayed engraftment following allogeneic hematopoietic stem cell transplantation. Cytotherapy 2024; 26:466-471. [PMID: 38430078 DOI: 10.1016/j.jcyt.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 11/08/2023] [Accepted: 01/25/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND AIMS Daratumumab, a human IgG monoclonal antibody targeting CD38, is a promising treatment for pediatric patients with relapsed or refractory T-cell acute lymphoblastic leukemia (T-ALL). We describe a case of delayed engraftment following a mismatched, unrelated donor hematopoietic stem cell transplant (HSCT) in a 14-year-old female with relapsed T-ALL, treated with daratumumab and chemotherapy. By Day 28 post-HSCT, the patient had no neutrophil engraftment but full donor myeloid chimerism. METHODS We developed two novel, semi-quantitative, antibody-based assays to measure the patient's bound and plasma daratumumab levels to determine if prolonged drug exposure may have contributed to her slow engraftment. RESULTS Daratumumab levels were significantly elevated more than 30 days after the patient's final infusion, and levels inversely correlated with her white blood cell counts. To clear daratumumab, the patient underwent several rounds of plasmapheresis and subsequently engrafted. CONCLUSIONS This is the first report of both delayed daratumumab clearance and delayed stem cell engraftment following daratumumab treatment in a pediatric patient. Further investigation is needed to elucidate the optimal dosing of daratumumab for treatment of acute leukemias in pediatric populations as well as daratumumab's potential effects on hematopoietic stem cells and stem cell engraftment following allogenic HSCT.
Collapse
Affiliation(s)
- Hannah Major-Monfried
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.
| | - Kinga Hosszu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Devin P McAvoy
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alexander Vallone
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alfred Gillio
- Pediatric Blood and Marrow Transplantation, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Barbara Spitzer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Pediatric Blood and Marrow Transplantation, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Andrew L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maria Cancio
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kevin Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andromachi Scaradavou
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joseph H Oved
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jaap Jan Boelens
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andrew C Harris
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
9
|
Lakkaraja M, Mauguen A, Boulad F, Cancio MI, Curran KJ, Harris AC, Kernan NA, Klein E, Kung AL, Oved J, Prockop S, Scaradavou A, Spitzer B, O'Reilly RJ, Boelens JJ. Impact of rabbit anti-thymocyte globulin (ATG) exposure on outcomes after ex vivo T-cell-depleted hematopoietic cell transplantation in pediatric and young adult patients. Cytotherapy 2024; 26:351-359. [PMID: 38349310 PMCID: PMC10997457 DOI: 10.1016/j.jcyt.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 11/03/2023] [Accepted: 01/24/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND AIMS Traditional weight-based dosing of rabbit anti-thymocyte globulin (rATG) used in allogeneic hematopoietic cell transplantation (HCT) to prevent graft-versus-host disease (GVHD) and graft rejection leads to variable exposures. High exposures induce delayed CD4+immune reconstitution (CD4+IR) and greater mortality. We sought to determine the impact of rATG exposure in children and young adults receiving various types of EX-VIVO T-cell-depleted (EX-VIVO-TCD) HCT. METHODS Patients receiving their first EX-VIVO-TCD HCT (CliniMACS CD34+, Isolex or soybean lectin agglutination), with removal of residual T cells by E-rosette depletion (E-) between 2008 and 2018 at Memorial Sloan Kettering Cancer Center were retrospectively analyzed. rATG exposure post-HCT was estimated (AU*d/L) using a validated population pharmacokinetic model. Previously defined rATG-exposures, <30, 30-55, ≥55 AU*d/L, were related with outcomes of interest. Cox proportional hazard and cause-specific models were used for analyses. RESULTS In total, 180 patients (median age 11 years; range 0.1-44 years) were included, malignant 124 (69%) and nonmalignant 56 (31%). Median post-HCT rATG exposure was 32 (0-104) AU*d/L. Exposure <30 AU*d/L was associated with a 3-fold greater probability of CD4+IR (P < 0.001); 2- to 4-fold lower risk of death (P = 0.002); and 3- to 4-fold lower risk of non-relapse mortality (NRM) (P = 0.02). Cumulative incidence of NRM was 8-fold lower in patients who attained CD4+IR compared with those who did not (P < 0.0001). There was no relation between rATG exposure and aGVHD (P = 0.33) or relapse (P = 0.23). Effect of rATG exposure on outcomes was similar in three EX-VIVO-TCD methods. CONCLUSIONS Individualizing rATG dosing to target a low rATG exposure post-HCT while maintaining total cumulative exposure may better predict CD4+IR, reduce NRM and increase overall survival, independent of the EX-VIVO-TCD method.
Collapse
Affiliation(s)
- Madhavi Lakkaraja
- Fred Hutchinson Cancer Center, Seattle, Washington, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Farid Boulad
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maria I Cancio
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Kevin J Curran
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Andrew C Harris
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Nancy A Kernan
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elizabeth Klein
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andrew L Kung
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joseph Oved
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Susan Prockop
- Dana Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andromachi Scaradavou
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Barbara Spitzer
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Richard J O'Reilly
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Jaap Jan Boelens
- Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
10
|
Borrill R, Poulton K, Wynn R. Immunology of cord blood T-cells favors augmented disease response during clinical pediatric stem cell transplantation for acute leukemia. Front Pediatr 2023; 11:1232281. [PMID: 37780051 PMCID: PMC10534014 DOI: 10.3389/fped.2023.1232281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/22/2023] [Indexed: 10/03/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) has been an important and efficacious treatment for acute leukemia in children for over 60 years. It works primarily through the graft-vs.-leukemia (GVL) effect, in which donor T-cells and other immune cells act to eliminate residual leukemia. Cord blood is an alternative source of stem cells for transplantation, with distinct biological and immunological characteristics. Retrospective clinical studies report superior relapse rates with cord blood transplantation (CBT), when compared to other stem cell sources, particularly for patients with high-risk leukemia. Xenograft models also support the superiority of cord blood T-cells in eradicating malignancy, when compared to those derived from peripheral blood. Conversely, CBT has historically been associated with an increased risk of transplant-related mortality (TRM) and morbidity, particularly from infection. Here we discuss clinical aspects of CBT, the unique immunology of cord blood T-cells, their role in the GVL effect and future methods to maximize their utility in cellular therapies for leukemia, honing and harnessing their antitumor properties whilst managing the risks of TRM.
Collapse
Affiliation(s)
- Roisin Borrill
- Blood and Marrow Transplant Unit, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kay Poulton
- Transplantation Laboratory, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Robert Wynn
- Blood and Marrow Transplant Unit, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
11
|
Zhang W, Rowntree LC, Muttucumaru R, Damelang T, Aban M, Hurt AC, Auladell M, Esterbauer R, Wines B, Hogarth M, Turner SJ, Wheatley AK, Kent SJ, Patil S, Avery S, Morrissey O, Chung AW, Koutsakos M, Nguyen THO, Cheng AC, Kotsimbos TC, Kedzierska K. Robust immunity to influenza vaccination in haematopoietic stem cell transplant recipients following reconstitution of humoral and adaptive immunity. Clin Transl Immunology 2023; 12:e1456. [PMID: 37383182 PMCID: PMC10294294 DOI: 10.1002/cti2.1456] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 05/22/2023] [Accepted: 06/09/2023] [Indexed: 06/30/2023] Open
Abstract
Objectives Influenza causes significant morbidity and mortality, especially in high-risk populations. Although current vaccination regimens are the best method to combat annual influenza disease, vaccine efficacy can be low in high-risk groups, such as haematopoietic stem cell transplant (HSCT) recipients. Methods We comprehensively assessed humoral immunity, antibody landscapes, systems serology and influenza-specific B-cell responses, together with their phenotypes and isotypes, to the inactivated influenza vaccine (IIV) in HSCT recipients in comparison to healthy controls. Results Inactivated influenza vaccine significantly increased haemagglutination inhibition (HAI) titres in HSCT recipients, similar to healthy controls. Systems serology revealed increased IgG1 and IgG3 antibody levels towards the haemagglutinin (HA) head, but not to neuraminidase, nucleoprotein or HA stem. IIV also increased frequencies of total, IgG class-switched and CD21loCD27+ influenza-specific B cells, determined by HA probes and flow cytometry. Strikingly, 40% of HSCT recipients had markedly higher antibody responses towards A/H3N2 vaccine strain than healthy controls and showed cross-reactivity to antigenically drifted A/H3N2 strains by antibody landscape analysis. These superior humoral responses were associated with a greater time interval after HSCT, while multivariant analyses revealed the importance of pre-existing immune memory. Conversely, in HSCT recipients who did not respond to the first dose, the second IIV dose did not greatly improve their humoral response, although 50% of second-dose patients reached a seroprotective HAI titre for at least one of vaccine strains. Conclusions Our study demonstrates efficient, although time-dependent, immune responses to IIV in HSCT recipients, and provides insights into influenza vaccination strategies targeted to immunocompromised high-risk groups.
Collapse
Affiliation(s)
- Wuji Zhang
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Louise C Rowntree
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | | | - Timon Damelang
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Malet Aban
- World Health Organisation (WHO) Collaborating Centre for Reference and Research on Influenza, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Aeron C Hurt
- World Health Organisation (WHO) Collaborating Centre for Reference and Research on Influenza, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
- Product Development Medical Affairs, Infectious DiseasesF. Hoffmann-La Roche LtdBaselSwitzerland
| | - Maria Auladell
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Robyn Esterbauer
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | | | | | - Stephen J Turner
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, and Department of MicrobiologyMonash UniversityClaytonVICAustralia
| | - Adam K Wheatley
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Stephen J Kent
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
- Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Sushrut Patil
- Malignant Haematology and Stem Cell Transplantation Service, Department of Clinical HaematologyThe Alfred HospitalMelbourneVICAustralia
| | - Sharon Avery
- Malignant Haematology and Stem Cell Transplantation Service, Department of Clinical HaematologyThe Alfred HospitalMelbourneVICAustralia
| | - Orla Morrissey
- Department of Infectious DiseasesAlfred HealthMelbourneVICAustralia
| | - Amy W Chung
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Marios Koutsakos
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Thi HO Nguyen
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Allen C Cheng
- School of Public Health and Preventive MedicineMonash UniversityClaytonVICAustralia
- Infection Prevention and Healthcare Epidemiology UnitAlfred HealthMelbourneVICAustralia
| | - Tom C Kotsimbos
- Department of Respiratory MedicineThe Alfred HospitalMelbourneVICAustralia
- Department of Medicine, Central Clinical School, The Alfred HospitalMonash UniversityMelbourneVICAustralia
| | - Katherine Kedzierska
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI‐CoRE)Hokkaido UniversitySapporoJapan
| |
Collapse
|
12
|
Spadea M, Saglio F, Ceolin V, Barone M, Zucchetti G, Quarello P, Fagioli F. Immune-mediated cytopenias (IMCs) after HSCT for pediatric non-malignant disorders: epidemiology, risk factors, pathogenesis, and treatment. Eur J Pediatr 2023; 182:2471-2483. [PMID: 36967419 PMCID: PMC10257634 DOI: 10.1007/s00431-023-04912-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 06/11/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) represents a curative option for pediatric patients affected by malignant and non-malignant disorders. Several complications may arise during the post-transplantation period, including immune-mediated disorders. Immune-mediated cytopenias (IMCs) account for up to 22% of pediatric HSCT complications, representing an important cause of morbidity and mortality post-HSCT. So far, their pathogenesis is not well-understood, and their management may be very challenging. Further, most patients are refractory to first-line treatment which is based on high-dose intravenous steroids, immunoglobulin, and the monoclonal anti-CD20 antibody - rituximab. No clear consensus has been reached for second- and third-line therapeutic options. CONCLUSION We reviewed the epidemiology, risk factors, pathogenesis, and treatment of IMCs, aiming to offer a deeper understanding of these complications as a guide to improving the management of these fragile patients and a cue for the design of tailored clinical trials. WHAT IS KNOWN • IMCs arising in the post-HSCT setting represent a rare but potentially life-threatening complication. Younger patients affected by non-malignant disorders are at the greatest risk of IMCs arising after HSCT. Corticosteroids, intravenous immunoglobulin, and rituximab represent the undiscussed first-line therapeutic approach. WHAT IS NEW • This review highlitghts how children present unique risk factors for post HSCT IMCs, which are the result of the complex relationship between the immaturity of their infantile immune system and all the perturbing agents and factors which characterize the post-HSCT setting. Future efforts are warranted to establish the best option for refractory patients, for whom a standard and validated approach is not currently available. Among new agents, ibrutinib or bortezomib and fostamatinib or low-dose IL-2 could represent a good therapeutic option for patients with graft-versus-host disease and hemolytic anemia or graft-versus-host disease and thrombocytopenia, respectively.
Collapse
Affiliation(s)
- Manuela Spadea
- Stem Cell Transplantation and Cellular Therapy Department, Pediatric Onco-Hematology, Azienda Ospedaliera-Universitaria Città Della Salute E Della Scienza, Regina Margherita Children's Hospital, Turin, Italy
- University of Torino, Turin, Italy
| | - Francesco Saglio
- Stem Cell Transplantation and Cellular Therapy Department, Pediatric Onco-Hematology, Azienda Ospedaliera-Universitaria Città Della Salute E Della Scienza, Regina Margherita Children's Hospital, Turin, Italy
| | - Valeria Ceolin
- Stem Cell Transplantation and Cellular Therapy Department, Pediatric Onco-Hematology, Azienda Ospedaliera-Universitaria Città Della Salute E Della Scienza, Regina Margherita Children's Hospital, Turin, Italy
- Erasmus University MC-Sophia Childrens Hospital, Rotterdam, Netherlands
| | - Marta Barone
- Stem Cell Transplantation and Cellular Therapy Department, Pediatric Onco-Hematology, Azienda Ospedaliera-Universitaria Città Della Salute E Della Scienza, Regina Margherita Children's Hospital, Turin, Italy
| | - Giulia Zucchetti
- Stem Cell Transplantation and Cellular Therapy Department, Pediatric Onco-Hematology, Azienda Ospedaliera-Universitaria Città Della Salute E Della Scienza, Regina Margherita Children's Hospital, Turin, Italy
| | - Paola Quarello
- Stem Cell Transplantation and Cellular Therapy Department, Pediatric Onco-Hematology, Azienda Ospedaliera-Universitaria Città Della Salute E Della Scienza, Regina Margherita Children's Hospital, Turin, Italy.
- University of Torino, Turin, Italy.
| | - Franca Fagioli
- Stem Cell Transplantation and Cellular Therapy Department, Pediatric Onco-Hematology, Azienda Ospedaliera-Universitaria Città Della Salute E Della Scienza, Regina Margherita Children's Hospital, Turin, Italy
- University of Torino, Turin, Italy
| |
Collapse
|
13
|
Lei YY, Chen XR, Jiang S, Guo M, Yu CL, Qiao JH, Cai B, Ai HS, Wang Y, Hu KX. Mechanisms of thymic repair of in vitro-induced precursor T cells as a haplo-identical HSCT regimen. Transplant Cell Ther 2023:S2666-6367(23)01174-0. [PMID: 36944387 DOI: 10.1016/j.jtct.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/06/2023] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) is currently an effective treatment for malignant hematological disease, but the immune deficiency and severe infection triggered by slow immune reconstitution are the main causes of high mortality and transplant failure. One of these outstanding problems is thymus damage, which is associated with graft-versus-host disease (GVHD), and preconditioning including irradiation and chemotherapy. Therefore, rapid repair of damaged thymus and rapid proliferation of thymus-derived donor T cells after transplantation are key to solving the problem. This study is designed to accelerate the recovery of thymus-derived T cells after transplantation. Wild-type mice with normal immunity were used as recipients in a haplo-HSCT mouse model to mimic clinical haplo-HSCT. A modified cell culture system using Notch ligand Delta4 and IL-7 was established that is capable of inducing and amplifying the differentiation and proliferation of hematopoietic stem cells into precursor T (preT) cells in vitro. Haplo-HSCT protocol included the preT and G-CSF mobilized donor splenic mononuclear cells (MNC) co-infusion or MNC alone. Thymic GVHD, thymic repair, and thymus-derived T cell development were compared in two groups by polychromatic immunofluorescence tracking, flow cytometry and detection of T cell receptor Vβ. The thymus homing and T-cell regeneration of allogenic preT cells were observed. The functions of preT cells in accelerating immune reconstitution, restoring thymic architecture, weakening GVH effects, and enhancing immuno-tolerance after transplantation were demonstrated. Further studies revealed that allogeneic preT cells induced by a culture system containing IL7 and Delta4 highly express ccr9 and RANKL. Interestingly, the RANK expression was promoted after preT cells' thymus homing. These results suggested that the RANK/RANKL pathway may play an important role in thymus homing. Our results provide a potential therapeutic option to optimize haplo-HSCT. It further opened up a new field of T cell therapy for artificial induction of allogeneic precursor T cells in vitro to repair the damaged thymus from irradiation and chemotherapy, and to compensate for the recovery of immune function in patients with immune deficiency caused by multiple reasons.
Collapse
Affiliation(s)
- Yang-Yang Lei
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China.
| | - Xin-Rui Chen
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Shan Jiang
- Anhui medical university, anhui province, China
| | - Mei Guo
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Chang-Lin Yu
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Jian-Hui Qiao
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Bo Cai
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Hui-Sheng Ai
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China
| | - Yi Wang
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China.
| | - Kai-Xun Hu
- Department of Hematology and Transplantation, the Fifth medical center, General Hospital of the People's Liberation Army, Beijing, China.
| |
Collapse
|
14
|
Donor natural killer cells trigger production of β-2-microglobulin to enhance post-bone marrow transplant immunity. Blood 2022; 140:2323-2334. [PMID: 35984965 DOI: 10.1182/blood.2021015297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 07/14/2022] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic transplantation is a powerful treatment for hematologic malignancies. Posttransplant immune incompetence exposes patients to disease relapse and infections. We previously demonstrated that donor alloreactive natural killer (NK) cells ablate recipient hematopoietic targets, including leukemia. Here, in murine models, we show that infusion of donor alloreactive NK cells triggers recipient dendritic cells (DCs) to synthesize β-2-microglobulin (B2M) that elicits the release of c-KIT ligand and interleukin-7 that greatly accelerate posttransplant immune reconstitution. An identical chain of events was reproduced by infusing supernatants of alloreactive NK/DC cocultures. Similarly, human alloreactive NK cells triggered human DCs to synthesize B2M that induced interleukin-7 production by thymic epithelial cells and thereby supported thymocyte cellularity in vitro. Chromatography fractionation of murine and human alloreactive NK/DC coculture supernatants identified a protein with molecular weight and isoelectric point of B2M, and mass spectrometry identified amino acid sequences specific of B2M. Anti-B2M antibody depletion of NK/DC coculture supernatants abrogated their immune-rebuilding effect. B2M knock-out mice were unable to undergo accelerated immune reconstitution, but infusion of (wild-type) NK/DC coculture supernatants restored their ability to undergo accelerated immune reconstitution. Similarly, silencing the B2M gene in human DCs, before coculture with alloreactive NK cells, prevented the increase in thymocyte cellularity in vitro. Finally, human recombinant B2M increased thymocyte cellularity in a thymic epithelial cells/thymocyte culture system. Our studies uncover a novel therapeutic principle for treating posttransplant immune incompetence and suggest that, upon its translation to the clinic, patients may benefit from adoptive transfer of large numbers of cytokine-activated, ex vivo-expanded donor alloreactive NK cells.
Collapse
|
15
|
Waller EK. Donor NK cells facilitate thymopoiesis in allo-BMT. Blood 2022; 140:2307-2308. [PMID: 36454592 DOI: 10.1182/blood.2022017856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
16
|
Gaballa A, Arruda LCM, Uhlin M. Gamma delta T-cell reconstitution after allogeneic HCT: A platform for cell therapy. Front Immunol 2022; 13:971709. [PMID: 36105821 PMCID: PMC9465162 DOI: 10.3389/fimmu.2022.971709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Allogeneic Hematopoietic stem cell transplantation (allo-HCT) is a curative platform for several hematological diseases. Despite its therapeutic benefits, the profound immunodeficiency associated with the transplant procedure remains a major challenge that renders patients vulnerable to several complications. Today, It is well established that a rapid and efficient immune reconstitution, particularly of the T cell compartment is pivotal to both a short-term and a long-term favorable outcome. T cells expressing a TCR heterodimer comprised of gamma (γ) and delta (δ) chains have received particular attention in allo-HCT setting, as a large body of evidence has indicated that γδ T cells can exert favorable potent anti-tumor effects without inducing severe graft versus host disease (GVHD). However, despite their potential role in allo-HCT, studies investigating their detailed reconstitution in patients after allo-HCT are scarce. In this review we aim to shed lights on the current literature and understanding of γδ T cell reconstitution kinetics as well as the different transplant-related factors that may influence γδ reconstitution in allo-HCT. Furthermore, we will present data from available reports supporting a role of γδ cells and their subsets in patient outcome. Finally, we discuss the current and future strategies to develop γδ cell-based therapies to exploit the full immunotherapeutic potential of γδ cells in HCT setting.
Collapse
Affiliation(s)
- Ahmed Gaballa
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Chemistry, National Liver Institute, Menoufia University, Menoufia, Egypt
| | - Lucas C. M. Arruda
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Michael Uhlin
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
17
|
Sottoriva K, Paik NY, White Z, Bandara T, Shao L, Sano T, Pajcini KV. A Notch/IL-21 signaling axis primes bone marrow T cell progenitor expansion. JCI Insight 2022; 7:e157015. [PMID: 35349492 PMCID: PMC9090257 DOI: 10.1172/jci.insight.157015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
Long-term impairment in T cell-mediated adaptive immunity is a major clinical obstacle following treatment of blood disorders with hematopoietic stem cell transplantation. Although T cell development in the thymus has been extensively characterized, there are significant gaps in our understanding of prethymic processes that influence early T cell potential. We have uncovered a Notch/IL-21 signaling axis in bone marrow common lymphoid progenitor (CLP) cells. IL-21 receptor expression was driven by Notch activation in CLPs, and in vivo treatment with IL-21 induced Notch-dependent CLP proliferation. Taking advantage of this potentially novel signaling axis, we generated T cell progenitors ex vivo, which improved repopulation of the thymus and peripheral lymphoid organs of mice in an allogeneic transplant model. Importantly, Notch and IL-21 activation were equally effective in the priming and expansion of human cord blood cells toward the T cell fate, confirming the translational potential of the combined treatment.
Collapse
Affiliation(s)
| | - Na Yoon Paik
- Department of Pharmacology and Regenerative Medicine and
| | - Zachary White
- Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | | | - Lijian Shao
- Department of Pharmacology and Regenerative Medicine and
| | - Teruyuki Sano
- Department of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | | |
Collapse
|
18
|
Jarduli-Maciel LR, de Azevedo JTC, Clave E, Costa TCDM, Arruda LCM, Fournier I, Palma PVB, Lima KC, Elias JB, Stracieri ABP, Pieroni F, Cunha R, Darrigo-Júnior LG, Grecco CES, Covas DT, Silva-Pinto AC, De Santis GC, Simões BP, Oliveira MC, Toubert A, Malmegrim KCR. Allogeneic haematopoietic stem cell transplantation resets T- and B-cell compartments in sickle cell disease patients. Clin Transl Immunology 2022; 11:e1389. [PMID: 35474905 PMCID: PMC9035210 DOI: 10.1002/cti2.1389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/12/2022] Open
Abstract
Objectives Allogeneic haematopoietic stem cell transplantation (allo-HSCT) is the only currently available curative treatment for sickle cell disease (SCD). Here, we comprehensively evaluated the reconstitution of T- and B-cell compartments in 29 SCD patients treated with allo-HSCT and how it correlated with the development of acute graft-versus-host disease (aGvHD). Methods T-cell neogenesis was assessed by quantification of signal-joint and β-chain TCR excision circles. B-cell neogenesis was evaluated by quantification of signal-joint and coding-joint K-chain recombination excision circles. T- and B-cell peripheral subset numbers were assessed by flow cytometry. Results Before allo-HSCT (baseline), T-cell neogenesis was normal in SCD patients compared with age-, gender- and ethnicity-matched healthy controls. Following allo-HSCT, T-cell neogenesis declined but was fully restored to healthy control levels at one year post-transplantation. Peripheral T-cell subset counts were fully restored only at 24 months post-transplantation. Occurrence of acute graft-versus-host disease (aGvHD) transiently affected T- and B-cell neogenesis and overall reconstitution of T- and B-cell peripheral subsets. B-cell neogenesis was significantly higher in SCD patients at baseline than in healthy controls, remaining high throughout the follow-up after allo-HSCT. Notably, after transplantation SCD patients showed increased frequencies of IL-10-producing B-regulatory cells and IgM+ memory B-cell subsets compared with baseline levels and with healthy controls. Conclusion Our findings revealed that the T- and B-cell compartments were normally reconstituted in SCD patients after allo-HSCT. In addition, the increase of IL-10-producing B-regulatory cells may contribute to improve immune regulation and homeostasis after transplantation.
Collapse
Affiliation(s)
- Luciana Ribeiro Jarduli-Maciel
- Graduate Program in Biosciences Applied to Pharmacy School of Pharmaceutical Sciences of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | - Júlia Teixeira Cottas de Azevedo
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Graduate Program in Basic and Applied Immunology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | | | - Thalita Cristina de Mello Costa
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,University Hospital of Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | | | - Isabelle Fournier
- Laboratoire d'Immunologie et d'Histocompatibilité Hôpital Saint-Louis AP-HP Paris France
| | - Patrícia Vianna Bonini Palma
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | - Keli Cristina Lima
- Graduate Program in Biosciences Applied to Pharmacy School of Pharmaceutical Sciences of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| | | | | | - Fabiano Pieroni
- Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Renato Cunha
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | | | | | - Dimas Tadeu Covas
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Ana Cristina Silva-Pinto
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,University Hospital of Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Gil Cunha De Santis
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,University Hospital of Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Belinda Pinto Simões
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Maria Carolina Oliveira
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Ribeirão Preto Medical School University of São Paulo São Paulo SP Brazil
| | - Antoine Toubert
- Université de Paris INSERM UMR 1160 IRSL Paris France.,Laboratoire d'Immunologie et d'Histocompatibilité Hôpital Saint-Louis AP-HP Paris France
| | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-Based Therapy Regional Blood Center of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil.,Department of Clinical Analysis, Toxicology and Food Sciences School of Pharmaceutical Sciences of Ribeirão Preto University of São Paulo Ribeirão Preto SP Brazil
| |
Collapse
|
19
|
Jazbec K, Jež M, Švajger U, Smrekar B, Miceska S, Rajčevič U, Justin M, Završnik J, Malovrh T, Švara T, Gombač M, Ramšak Ž, Rožman P. The Influence of Heterochronic Non-Myeloablative Bone Marrow Transplantation on the Immune System, Frailty, General Health, and Longevity of Aged Murine Recipients. Biomolecules 2022; 12:biom12040595. [PMID: 35454183 PMCID: PMC9028083 DOI: 10.3390/biom12040595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/06/2022] [Accepted: 04/14/2022] [Indexed: 12/10/2022] Open
Abstract
The stem cell theory of aging postulates that stem cells become inefficient at maintaining the original functions of the tissues. We, therefore, hypothesized that transplanting young bone marrow (BM) to old recipients would lead to rejuvenating effects on immunity, followed by improved general health, decreased frailty, and possibly life span extension. We developed a murine model of non-myeloablative heterochronic BM transplantation in which old female BALB/c mice at 14, 16, and 18(19) months of age received altogether 125.1 ± 15.6 million nucleated BM cells from young male donors aged 7–13 weeks. At 21 months, donor chimerism was determined, and the immune system’s innate and adaptive arms were analyzed. Mice were then observed for general health and frailty until spontaneous death, when their lifespan, post-mortem examinations, and histopathological changes were recorded. The results showed that the old mice developed on average 18.7 ± 9.6% donor chimerism in the BM and showed certain improvements in their innate and adaptive arms of the immune system, such as favorable counts of neutrophils in the spleen and BM, central memory Th cells, effector/effector memory Th and Tc cells in the spleen, and B1a and B1b cells in the peritoneal cavity. Borderline enhanced lymphocyte proliferation capacity was also seen. The frailty parameters, pathomorphological results, and life spans did not differ significantly in the transplanted vs. control group of mice. In conclusion, although several favorable effects are obtained in our heterochronic non-myeloablative transplantation model, additional optimization is needed for better rejuvenation effects.
Collapse
Affiliation(s)
- Katerina Jazbec
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
- Correspondence:
| | - Mojca Jež
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Urban Švajger
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
- Chair of Clinical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Boštjan Smrekar
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Simona Miceska
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Uroš Rajčevič
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Mojca Justin
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Janja Završnik
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia;
| | - Tadej Malovrh
- Institute of Microbiology and Parasitology, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Tanja Švara
- Institute of Pathology, Wild Animals, Fish and Bees, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.Š.); (M.G.)
| | - Mitja Gombač
- Institute of Pathology, Wild Animals, Fish and Bees, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.Š.); (M.G.)
| | - Živa Ramšak
- National Institute of Biology, 1000 Ljubljana, Slovenia;
| | - Primož Rožman
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| |
Collapse
|
20
|
Kim YK, Lee JM. Change of RBC Deformability During Hematopoietic Stem Cell Transplantation. J Pediatr Hematol Oncol 2022; 44:e329-e333. [PMID: 34486554 DOI: 10.1097/mph.0000000000002295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/02/2021] [Indexed: 11/26/2022]
Abstract
The red blood cell (RBC) deformability test is the measurement of the ability of RBCs to adapt their shape to the flow conditions. The major determinants of RBC deformability include cell shape, composition of the cell membrane and cytoskeleton, and internal viscosity (mean cell hemoglobin concentration). RBC deformability is primarily regulated by the composition and arrangement of the cell membrane. In cancer patients, chemotherapy and hematopoietic stem transplantation (HSCT) affect the bone marrow microenvironment, which may alter RBC production and deformability. We aimed to evaluate the change in RBC deformability during HSCT. Blood samples were obtained from patients who underwent HSCT. Eleven children were enrolled in this study. RBC deformability was measured with a microfluidic ektacytometer (RheoScan-D, RheoMeditech, Seoul, Korea). All analyses were completed within 24 hours after blood collection. The elongation index of the erythrocytes was measured. The elongation index of RBCs gradually increased from day 5 to day 30 after HSCT. RBC deformability may reflect the bone marrow microenvironment of the patient during HSCT. Further studies investigating the correlation between RBC deformability and the prognosis of HSCT are needed.
Collapse
Affiliation(s)
- Yu Kyung Kim
- Department of Clinical Pathology, School of Medicine, Kyungpook National University
| | - Jae Min Lee
- Department of Pediatrics, Yeungnam University College of Medicine, Daegu, Republic of Korea
| |
Collapse
|
21
|
Alawam AS, Cosway EJ, James KD, Lucas B, Bacon A, Parnell SM, White AJ, Jenkinson WE, Anderson G. Failures in thymus medulla regeneration during immune recovery cause tolerance loss and prime recipients for auto-GVHD. J Exp Med 2022; 219:212911. [PMID: 34910105 PMCID: PMC8679781 DOI: 10.1084/jem.20211239] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/20/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022] Open
Abstract
Bone marrow transplantation (BMT) is a widely used therapy for blood cancers and primary immunodeficiency. Following transplant, the thymus plays a key role in immune reconstitution by generating a naive αβT cell pool from transplant-derived progenitors. While donor-derived thymopoiesis during the early post-transplant period is well studied, the ability of the thymus to synchronize T cell development with essential tolerance mechanisms is poorly understood. Using a syngeneic mouse transplant model, we analyzed T cell recovery alongside the regeneration and function of intrathymic microenvironments. We report a specific and prolonged failure in the post-transplant recovery of medullary thymic epithelial cells (mTECs). This manifests as loss of medulla-dependent tolerance mechanisms, including failures in Foxp3+ regulatory T cell development and formation of the intrathymic dendritic cell pool. In addition, defective negative selection enables escape of self-reactive conventional αβT cells that promote autoimmunity. Collectively, we show that post-transplant T cell recovery involves an uncoupling of thymopoiesis from thymic tolerance, which results in autoimmune reconstitution caused by failures in thymic medulla regeneration.
Collapse
Affiliation(s)
- Abdullah S Alawam
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Emilie J Cosway
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Kieran D James
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Beth Lucas
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Andrea Bacon
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Sonia M Parnell
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Andrea J White
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - William E Jenkinson
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Graham Anderson
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| |
Collapse
|
22
|
Early Reconstitution of Antibody Secreting Cells after Allogeneic Stem Cell Transplantation. J Clin Med 2022; 11:jcm11010270. [PMID: 35012014 PMCID: PMC8745805 DOI: 10.3390/jcm11010270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/26/2021] [Accepted: 01/03/2022] [Indexed: 11/24/2022] Open
Abstract
Immune cell reconstitution after stem cell transplantation is allocated over several stages. Whereas cells mediating innate immunity recover rapidly, adaptive immune cells, including T and B cells, recover slowly over several months. In this study we investigated kinetics and reconstitution of de novo B cell formation in patients receiving CD3 and CD19 depleted haploidentical stem cell transplantation with additional in vivo T cell depletion with monoclonal anti-CD3 antibody. This model enables a detailed in vivo evaluation of hierarchy and attribution of defined lymphocyte populations without skewing by mTOR- or NFAT-inhibitors. As expected CD3+ T cells and their subsets had delayed reconstitution (<100 cells/μL at day +90). Well defined CD19+ B lymphocytes of naïve and memory phenotype were detected at day +60. Remarkably, we observed a very early reconstitution of antibody-secreting cells (ASC) at day +14. These ASC carried the HLA-haplotype of the donor and secreted the isotypes IgM and IgA more prevalent than IgG. They correlated with a population of CD19− CD27− CD38low/+ CD138− cells. Of note, reconstitution of this ASC occurred without detectable circulating T cells and before increase of BAFF or other B cell stimulating factors. In summary, we describe a rapid reconstitution of peripheral blood ASC after CD3 and CD19 depleted haploidentical stem cell transplantation, far preceding detection of naïve and memory type B cells. Incidence before T cell reconstitution and spontaneous secretion of immunoglobulins allocate these early ASC to innate immunity, eventually maintaining natural antibody levels.
Collapse
|
23
|
Antithymocyte globulin exposure in CD34+ T-cell depleted allogeneic hematopoietic cell transplantation. Blood Adv 2021; 6:1054-1063. [PMID: 34788361 PMCID: PMC8945304 DOI: 10.1182/bloodadvances.2021005584] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022] Open
Abstract
In CD34+ TCD allogeneic-HCT, optimum post-HCT rATG exposure decreases NRM driven by faster CD4+ IR and improves survival. Personalized rATG exposure using a PK-directed strategy may improve survival after allogeneic CD34+ TCD HCT.
Traditional weight-based dosing results in variable rabbit antithymocyte globulin (rATG) clearance that can delay CD4+ T-cell immune reconstitution (CD4+ IR) leading to higher mortality. In a retrospective pharmacokinetic/pharmacodynamic (PK/PD) analysis of patients undergoing their first CD34+ T-cell–depleted (TCD) allogeneic hematopoietic cell transplantation (HCT) after myeloablative conditioning with rATG, we estimated post-HCT rATG exposure as area under the curve (arbitrary unit per day/milliliter [AU × day/mL]) using a validated population PK model. We related rATG exposure to nonrelapse mortality (NRM), CD4+ IR (CD4+ ≥50 cells per µL at 2 consecutive measures within 100 days after HCT), overall survival, relapse, and acute graft-versus-host disease (aGVHD) to define an optimal rATG exposure. We used Cox proportional hazard models and multistate competing risk models for analysis. In all, 554 patients were included (age range, 0.1-73 years). Median post-HCT rATG exposure was 47 AU × day/mL (range, 0-101 AU × day/mL). Low post-HCT area under the curve (<30 AU × day/mL) was associated with lower risk of NRM (P < .01) and higher probability of achieving CD4+ IR (P < .001). Patients who attained CD4+ IR had a sevenfold lower 5-year NRM (P < .0001). The probability of achieving CD4+ IR was 2.5-fold higher in the <30 AU × day/mL group compared with 30-55 AU × day/mL and threefold higher in the <30 AU × day/mL group compared with the ≥55 AU × day/mL group. In multivariable analyses, post-HCT rATG exposure ≥55 AU × day/mL was associated with an increased risk of NRM (hazard ratio, 3.42; 95% confidence interval, 1.26-9.30). In the malignancy subgroup (n = 515), a tenfold increased NRM was observed in the ≥55 AU × day/mL group, and a sevenfold increased NRM was observed in the 30-55 AU × day/mL group compared with the <30 AU × day/mL group. Post-HCT rATG exposure ≥55 AU × day/mL was associated with higher risk of a GVHD (hazard ratio, 2.28; 95% confidence interval, 1.01-5.16). High post-HCT rATG exposure is associated with higher NRM secondary to poor CD4+ IR after TCD HCT. Using personalized PK-directed rATG dosing to achieve optimal exposure may improve survival after HCT.
Collapse
|
24
|
Tourret M, Talvard-Balland N, Lambert M, Ben Youssef G, Chevalier MF, Bohineust A, Yvorra T, Morin F, Azarnoush S, Lantz O, Dalle JH, Caillat-Zucman S. Human MAIT cells are devoid of alloreactive potential: prompting their use as universal cells for adoptive immune therapy. J Immunother Cancer 2021; 9:jitc-2021-003123. [PMID: 34615705 PMCID: PMC8496386 DOI: 10.1136/jitc-2021-003123] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Background Mucosal-associated invariant T (MAIT) cells are semi-invariant T cells that recognize microbial antigens presented by the highly conserved MR1 molecule. MAIT cells are predominantly localized in the liver and barrier tissues and are potent effectors of antimicrobial defense. MAIT cells are very few at birth and accumulate gradually over a period of about 6 years during the infancy. The cytotoxic potential of MAIT cells, as well as their newly described regulatory and tissue repair functions, open the possibility of exploiting their properties in adoptive therapy. A prerequisite for their use as ‘universal’ cells would be a lack of alloreactive potential, which remains to be demonstrated. Methods We used ex vivo, in vitro and in vivo models to determine if human MAIT cells contribute to allogeneic responses. Results We show that recovery of MAIT cells after allogeneic hematopoietic stem cell transplantation recapitulates their slow physiological expansion in early childhood, independent of recovery of non-MAIT T cells. In vitro, signals provided by allogeneic cells and cytokines do not induce sustained MAIT cell proliferation. In vivo, human MAIT cells do not expand nor accumulate in tissues in a model of T-cell-mediated xenogeneic graft-versus-host disease in immunodeficient mice. Conclusions Altogether, these results provide evidence that MAIT cells are devoid of alloreactive potential and pave the way for harnessing their translational potential in universal adoptive therapy overcoming barriers of HLA disparity. Trial registration number ClinicalTrials.gov number NCT02403089.
Collapse
Affiliation(s)
- Marie Tourret
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Nana Talvard-Balland
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Marion Lambert
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Ghada Ben Youssef
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Mathieu F Chevalier
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Armelle Bohineust
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Thomas Yvorra
- INSERM UMR3666/U1143, Université PSL, Institut Curie, Paris, France
| | - Florence Morin
- Laboratoire d'Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France
| | - Saba Azarnoush
- Département d'Immuno-Hématologie, Hôpital Robert Debré, AP-HP, Université de Paris, Paris, France
| | - Olivier Lantz
- INSERM U932, Université PSL, Institut Curie, Paris, France.,Laboratoire d'immunologie clinique & Centre d'investigation Clinique en Biothérapie (CIC-BT1428), Institut Curie, Paris, France
| | - Jean-Hugues Dalle
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France.,Département d'Immuno-Hématologie, Hôpital Robert Debré, AP-HP, Université de Paris, Paris, France
| | - Sophie Caillat-Zucman
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France .,Laboratoire d'Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France
| |
Collapse
|
25
|
Targeting the actin nucleation promoting factor WASp provides a therapeutic approach for hematopoietic malignancies. Nat Commun 2021; 12:5581. [PMID: 34552085 PMCID: PMC8458504 DOI: 10.1038/s41467-021-25842-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/03/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer cells depend on actin cytoskeleton rearrangement to carry out hallmark malignant functions including activation, proliferation, migration and invasiveness. Wiskott–Aldrich Syndrome protein (WASp) is an actin nucleation-promoting factor and is a key regulator of actin polymerization in hematopoietic cells. The involvement of WASp in malignancies is incompletely understood. Since WASp is exclusively expressed in hematopoietic cells, we performed in silico screening to identify small molecule compounds (SMCs) that bind WASp and promote its degradation. We describe here one such identified molecule; this WASp-targeting SMC inhibits key WASp-dependent actin processes in several types of hematopoietic malignancies in vitro and in vivo without affecting naïve healthy cells. This small molecule demonstrates limited toxicity and immunogenic effects, and thus, might serve as an effective strategy to treat specific hematopoietic malignancies in a safe and precisely targeted manner. Cancer cells proliferate and invade via cytoskeletal proteins such as WASp, exclusively expressed in hematopoietic cells. Here the authors show a specific small molecule compound inhibiting cancer cell activity by WASp degradation and demonstrating its therapeutic potential in vitro and in vivo.
Collapse
|
26
|
Duah M, Li L, Shen J, Lan Q, Pan B, Xu K. Thymus Degeneration and Regeneration. Front Immunol 2021; 12:706244. [PMID: 34539637 PMCID: PMC8442952 DOI: 10.3389/fimmu.2021.706244] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/16/2021] [Indexed: 01/08/2023] Open
Abstract
The immune system’s ability to resist the invasion of foreign pathogens and the tolerance to self-antigens are primarily centered on the efficient functions of the various subsets of T lymphocytes. As the primary organ of thymopoiesis, the thymus performs a crucial role in generating a self-tolerant but diverse repertoire of T cell receptors and peripheral T cell pool, with the capacity to recognize a wide variety of antigens and for the surveillance of malignancies. However, cells in the thymus are fragile and sensitive to changes in the external environment and acute insults such as infections, chemo- and radiation-therapy, resulting in thymic injury and degeneration. Though the thymus has the capacity to self-regenerate, it is often insufficient to reconstitute an intact thymic function. Thymic dysfunction leads to an increased risk of opportunistic infections, tumor relapse, autoimmunity, and adverse clinical outcome. Thus, exploiting the mechanism of thymic regeneration would provide new therapeutic options for these settings. This review summarizes the thymus’s development, factors causing thymic injury, and the strategies for improving thymus regeneration.
Collapse
Affiliation(s)
- Maxwell Duah
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Lingling Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Jingyi Shen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Qiu Lan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Bin Pan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
27
|
Analysis of biological models to predict clinical outcomes based on HLA-DPB1 disparities in unrelated transplantation. Blood Adv 2021; 5:3377-3386. [PMID: 34448833 DOI: 10.1182/bloodadvances.2020003998] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/23/2021] [Indexed: 11/20/2022] Open
Abstract
HLA compatibility is a key factor for survival after unrelated hematopoietic stem cell transplantation (HSCT). HLA-A, -B, -C, -DRB1, and -DQB1 are usually matched between donor and recipient. By contrast, HLA-DPB1 mismatches are frequent, although it is feasible to optimize donor selection and DPB1 matching with prospective typing. Because classical DPB1 allele mismatches are often unavoidable, however, several biological models have been developed to predict the optimal DPB1 mismatch combination for less graft-versus-host disease (GVHD) and better overall survival. In 909 recipient/donor pairs, we analyzed the role of 3 biological models: T-cell epitopes (TCEs) based on the immunogenicity of DPB1, cell surface expression of DPB1 molecules based on a single-nucleotide polymorphism located in the 3' untranslated region, and the Predicted Indirectly ReCognizable HLA Epitopes (PIRCHE) model based on the presentation of allogeneic peptides derived from mismatched HLA, compared with the classical allele mismatch. Matching for both DPB1 alleles remains the best option to prevent acute GVHD. In the situation of one DPB1 allele mismatch, the donor associated with the lowest acute GVHD risks is mismatched for an allele with a low expression profile in the recipient, followed by a permissive TCE3/4 mismatch and/or the absence of PIRCHE II potential against the recipient. In the context of 2 DPB1 mismatches, the same considerations apply for a permissive TCE3/4 mismatch and no PIRCHE II. By combining the biological models, the most favorable DPB1 constellation can be defined. This approach will help optimize donor selection and improve post-HSCT complications and patient prognosis.
Collapse
|
28
|
Itamura H, Shindo T, Muranushi H, Kitaura K, Okada S, Shin-I T, Suzuki R, Takaori-Kondo A, Kimura S. Pharmacological MEK inhibition promotes polyclonal T-cell reconstitution and suppresses xenogeneic GVHD. Cell Immunol 2021; 367:104410. [PMID: 34274730 DOI: 10.1016/j.cellimm.2021.104410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 11/21/2022]
Abstract
Rapid immune reconstitution without developing graft-versus-host disease (GVHD) is required for the success of allogeneic hematopoietic stem cell transplantation. Here, we analyzed the effects of pharmacological MEK inhibition on human polyclonal T-cell reconstitution in a humanized mouse GVHD model utilizing deep sequencing-based T-cell receptor (TCR) repertoire analysis. GVHD mice exhibited a skewed TCR repertoire with a common clone within target organs. The MEK inhibitor trametinib ameliorated GVHD and enabled engraftment of diverse T-cell clones. Furthermore, trametinib also ameliorated GVHD sparing diverse T cell repertoire, even when it was given from day 15 through 28. Although tacrolimus also reduced development of GVHD, it disturbed diverse T cell reconstitution and resulted in skewed TCR repertoire. Thus, trametinib not only suppresses GVHD-inducing T cells but also promotes human T cell reconstitution in vivo, providing a novel rationale for translational studies targeting human GVHD.
Collapse
Affiliation(s)
- Hidekazu Itamura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Takero Shindo
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan; Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Hiroyuki Muranushi
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | | | - Ryuji Suzuki
- Repertoire Genesis Inc., Ibaraki, Japan; Department of Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, Sagamihara, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
29
|
Sarkkinen J, Lundgren S, Itälä‐Remes M, Salmenniemi U, Mustjoki S, Peterson P, Kekäläinen E. Anti‐cytokine autoantibodies are rare in chronic graft‐versus‐host disease. Scand J Immunol 2021. [DOI: 10.1111/sji.13091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Joona Sarkkinen
- Translational Immunology Research Program University of Helsinki Helsinki Finland
| | - Sofie Lundgren
- Translational Immunology Research Program University of Helsinki Helsinki Finland
- Hematology Research Unit Helsinki Department of Hematology University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center Helsinki Finland
| | - Maija Itälä‐Remes
- Department of Clinical Hematology and Stem Cell Transplantation Turku University Hospital and University of Turku Turku Finland
| | - Urpu Salmenniemi
- Department of Clinical Hematology and Stem Cell Transplantation Turku University Hospital and University of Turku Turku Finland
| | - Satu Mustjoki
- Translational Immunology Research Program University of Helsinki Helsinki Finland
- Hematology Research Unit Helsinki Department of Hematology University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center Helsinki Finland
- Department of Clinical Chemistry and Hematology University of Helsinki Helsinki Finland
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| | - Eliisa Kekäläinen
- Translational Immunology Research Program University of Helsinki Helsinki Finland
- HUSLAB Clinical Microbiology HUS Diagnostic Center Helsinki University Hospital Helsinki Finland
| |
Collapse
|
30
|
Tsilifis C, Moreira D, Marques L, Neves E, Slatter MA, Gennery AR. Stem cell transplantation as treatment for major histocompatibility class I deficiency. Clin Immunol 2021; 229:108801. [PMID: 34280577 DOI: 10.1016/j.clim.2021.108801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 11/17/2022]
Abstract
Major histocompatibility class I deficiency, due to genetic lesions in TAP1, TAP2, TAPBP, or B2M, manifests with recurrent sinopulmonary infections and granulomatous skin ulceration, and is predominately treated with antimicrobial prophylaxis and chest physiotherapy. One previous report of hematopoietic stem cell transplantation has been described in the literature, demonstrating cure of the immune defect without significant graft-versus-host disease. In this report, we expand the literature on HSCT in MHC-I deficiency with follow-up of the original patient, demonstrating maintained resolution of normal immune function and regression of the granulomatous rash 15 years post-transplant, and describe a further patient with mycobacterial disease whose transplant course was complicated by severe graft-versus-host disease.
Collapse
Affiliation(s)
- Christo Tsilifis
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital (GNCH), Victoria Wing, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, United Kingdom; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom.
| | - Diana Moreira
- Paediatric Infectious Diseases and Immunodeficiencies Unit and Clinical Immunology Unit, Centro Hospitalar e Universitário do Porto, Porto, Portugal
| | - Laura Marques
- Paediatric Infectious Diseases and Immunodeficiencies Unit and Clinical Immunology Unit, Centro Hospitalar e Universitário do Porto, Porto, Portugal
| | - Esmeralda Neves
- Immunology Service and Clinical Immunology Unit, Centro Hospitalar e Universitário do Porto, Unit for Multidisciplinary Research in Biomedicine, Porto, Portugal
| | - Mary A Slatter
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital (GNCH), Victoria Wing, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, United Kingdom; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Andrew R Gennery
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital (GNCH), Victoria Wing, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, United Kingdom; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| |
Collapse
|
31
|
Moirangthem RD, Ma K, Lizot S, Cordesse A, Olivré J, de Chappedelaine C, Joshi A, Cieslak A, Tchen J, Cagnard N, Asnafi V, Rausell A, Simons L, Zuber J, Taghon T, Staal FJT, Pflumio F, Six E, Cavazzana M, Lagresle-Peyrou C, Soheili T, André I. A DL-4- and TNFα-based culture system to generate high numbers of nonmodified or genetically modified immunotherapeutic human T-lymphoid progenitors. Cell Mol Immunol 2021; 18:1662-1676. [PMID: 34117371 PMCID: PMC8245454 DOI: 10.1038/s41423-021-00706-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
Several obstacles to the production, expansion and genetic modification of immunotherapeutic T cells in vitro have restricted the widespread use of T-cell immunotherapy. In the context of HSCT, delayed naïve T-cell recovery contributes to poor outcomes. A novel approach to overcome the major limitations of both T-cell immunotherapy and HSCT would be to transplant human T-lymphoid progenitors (HTLPs), allowing reconstitution of a fully functional naïve T-cell pool in the patient thymus. However, it is challenging to produce HTLPs in the high numbers required to meet clinical needs. Here, we found that adding tumor necrosis factor alpha (TNFα) to a DL-4-based culture system led to the generation of a large number of nonmodified or genetically modified HTLPs possessing highly efficient in vitro and in vivo T-cell potential from either CB HSPCs or mPB HSPCs through accelerated T-cell differentiation and enhanced HTLP cell cycling and survival. This study provides a clinically suitable cell culture platform to generate high numbers of clinically potent nonmodified or genetically modified HTLPs for accelerating immune recovery after HSCT and for T-cell-based immunotherapy (including CAR T-cell therapy).
Collapse
Affiliation(s)
- Ranjita Devi Moirangthem
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Kuiying Ma
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Sabrina Lizot
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Anne Cordesse
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Juliette Olivré
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Corinne de Chappedelaine
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Akshay Joshi
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Agata Cieslak
- grid.412134.10000 0004 0593 9113Laboratory of Onco-Hematology, AP-HP, Hôpital Necker-Enfants Malades., Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Institut Necker-Enfants Malades (INEM), INSERM UMR 1151, Paris, France
| | - John Tchen
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Nicolas Cagnard
- grid.508487.60000 0004 7885 7602Plateforme Bio-informatique, Université Paris Descartes, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS 3633, Paris, France
| | - Vahid Asnafi
- grid.412134.10000 0004 0593 9113Laboratory of Onco-Hematology, AP-HP, Hôpital Necker-Enfants Malades., Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Institut Necker-Enfants Malades (INEM), INSERM UMR 1151, Paris, France
| | - Antonio Rausell
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Clinical Bioinformatics, INSERM UMR 1163, Paris, France
| | - Laura Simons
- grid.412134.10000 0004 0593 9113Department of Biotherapy Clinical Investigation Center, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Julien Zuber
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France ,grid.412134.10000 0004 0593 9113Department of Adult Kidney Transplantation, AP-HP, Hôpital Necker, Paris, France
| | - Tom Taghon
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium ,grid.5342.00000 0001 2069 7798Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Frank J. T. Staal
- grid.10419.3d0000000089452978Department of Immunohematology & Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Françoise Pflumio
- grid.7429.80000000121866389Team Niche and Cancer in Hematopoiesis, Université de Paris and Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Emmanuelle Six
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Marina Cavazzana
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France ,grid.412134.10000 0004 0593 9113Department of Biotherapy Clinical Investigation Center, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Chantal Lagresle-Peyrou
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France ,grid.412134.10000 0004 0593 9113Department of Biotherapy Clinical Investigation Center, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Tayebeh Soheili
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| | - Isabelle André
- grid.508487.60000 0004 7885 7602Université de Paris, Imagine Institute, Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Paris, France
| |
Collapse
|
32
|
Gabelli M, Ademokun C, Cooper N, Amrolia PI. Pathogenesis, risk factors and therapeutic options for autoimmune haemolytic anaemia in the post-transplant setting. Br J Haematol 2021; 196:45-62. [PMID: 34195990 DOI: 10.1111/bjh.17648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/26/2021] [Indexed: 11/27/2022]
Abstract
Autoimmune haemolytic anaemia (AIHA) is a rare complication of allogeneic haematopoietic stem cell transplantation (HSCT), observed with an incidence of 1-5%. Paediatric age, diagnosis of non-malignant disease, lympho-depleting agents in the conditioning regimen, use of unrelated donor, graft versus host disease and infections have been associated with a higher risk of AIHA post HSCT. Post-HSCT AIHA is associated with high mortality and morbidity, and it is often very difficult to treat. Steroids and rituximab are used with a response rate around 30-50%. These and other therapeutic strategies are mainly derived from data on primary AIHA, although response rates in post-HSCT AIHA have been generally lower. Here we review the currently available data on risk factors and therapeutic options. There is a need for prospective studies in post-HSCT AIHA to guide clinicians in managing these complex patients.
Collapse
Affiliation(s)
- Maria Gabelli
- Department of Bone Marrow Transplantation, Great Ormond Street Hospital, London, UK
| | - Christine Ademokun
- Department of Haematology, Imperial College Healthcare NHS Trust, London, UK
| | - Nichola Cooper
- Department of Immunology and Inflammation, Imperial College, London, UK
| | - Persis I Amrolia
- Department of Bone Marrow Transplantation, Great Ormond Street Hospital, London, UK
| |
Collapse
|
33
|
Eberhard JM, Angin M, Passaes C, Salgado M, Monceaux V, Knops E, Kobbe G, Jensen B, Christopeit M, Kröger N, Vandekerckhove L, Badiola J, Bandera A, Raj K, van Lunzen J, Hütter G, Kuball JHE, Martinez-Laperche C, Balsalobre P, Kwon M, Díez-Martín JL, Nijhuis M, Wensing A, Martinez-Picado J, Schulze Zur Wiesch J, Sáez-Cirión A. Vulnerability to reservoir reseeding due to high immune activation after allogeneic hematopoietic stem cell transplantation in individuals with HIV-1. Sci Transl Med 2021; 12:12/542/eaay9355. [PMID: 32376772 DOI: 10.1126/scitranslmed.aay9355] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only medical intervention that has led to an HIV cure. Whereas the HIV reservoir sharply decreases after allo-HSCT, the dynamics of the T cell reconstitution has not been comprehensively described. We analyzed the activation and differentiation of CD4+ and CD8+ T cells, and the breadth and quality of HIV- and CMV-specific CD8+ T cell responses in 16 patients with HIV who underwent allo-HSCT (including five individuals who received cells from CCR5Δ32/Δ32 donors) to treat their underlying hematological malignancy and who remained on antiretroviral therapy (ART). We found that reconstitution of the T cell compartment after allo-HSCT was slow and heterogeneous with an initial expansion of activated CD4+ T cells that preceded the expansion of CD8+ T cells. Although HIV-specific CD8+ T cells disappeared immediately after allo-HSCT, weak HIV-specific CD8+ T cell responses were detectable several weeks after transplant and could still be detected at the time of full T cell chimerism, indicating that de novo priming, and hence antigen exposure, occurred during the time of T cell expansion. These HIV-specific T cells had limited functionality compared with CMV-specific CD8+ T cells and persisted years after allo-HSCT. In conclusion, immune reconstitution was slow, heterogeneous, and incomplete and coincided with de novo detection of weak HIV-specific T cell responses. The initial short phase of high T cell activation, in which HIV antigens were present, may constitute a window of vulnerability for the reseeding of viral reservoirs, emphasizing the importance of maintaining ART directly after allo-HSCT.
Collapse
Affiliation(s)
- Johanna M Eberhard
- 1. Department of Medicine, Infectious Diseases Unit, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany.,DZIF Partner Site (German Center for Infection Research), Hamburg-Lübeck-Borstel-Riems Site, Hamburg, Germany
| | - Mathieu Angin
- Institut Pasteur, HIV, Inflammation and Persistence, 75015 Paris, France
| | - Caroline Passaes
- Institut Pasteur, HIV, Inflammation and Persistence, 75015 Paris, France
| | - Maria Salgado
- AIDS Research Institute IrsiCaixa, 08916 Badalona, Spain
| | - Valerie Monceaux
- Institut Pasteur, HIV, Inflammation and Persistence, 75015 Paris, France
| | - Elena Knops
- Institute of Virology, University of Cologne, 50935 Cologne, Germany
| | - Guido Kobbe
- Department of Haematology, Oncology, and Clinical Immunology, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Björn Jensen
- Department of Gastroenterology, Hepatology, and Infectious Diseases, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Maximilian Christopeit
- Department of Stem Cell Transplantation, University Medical Center HamburgEppendorf, 20246 Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center HamburgEppendorf, 20246 Hamburg, Germany
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University and Ghent University Hospital, B-9000 Ghent, Belgium
| | - Jon Badiola
- Hematology Department, Virgen de las Nieves University Hospital, 18014 Granada, Spain
| | | | - Kavita Raj
- Department of Haematology, King's College Hospital, London SE5 9RS, UK
| | - Jan van Lunzen
- 1. Department of Medicine, Infectious Diseases Unit, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany.,ViiV Healthcare, Brentford, Middlesex TW8 9GS, UK
| | | | | | - Carolina Martinez-Laperche
- Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitarias Gregorio Marañón, Universidad Complutense, 28007 Madrid, Spain
| | - Pascual Balsalobre
- Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitarias Gregorio Marañón, Universidad Complutense, 28007 Madrid, Spain
| | - Mi Kwon
- Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitarias Gregorio Marañón, Universidad Complutense, 28007 Madrid, Spain
| | - José L Díez-Martín
- Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitarias Gregorio Marañón, Universidad Complutense, 28007 Madrid, Spain
| | - Monique Nijhuis
- University Medical Center Utrecht, 3584 CX, Utrecht, Netherlands
| | | | - Javier Martinez-Picado
- AIDS Research Institute IrsiCaixa, 08916 Badalona, Spain.,UVic-UCC, 08500 Vic, Spain.,ICREA, 08010 Barcelona, Spain
| | - Julian Schulze Zur Wiesch
- 1. Department of Medicine, Infectious Diseases Unit, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany. .,DZIF Partner Site (German Center for Infection Research), Hamburg-Lübeck-Borstel-Riems Site, Hamburg, Germany
| | - Asier Sáez-Cirión
- Institut Pasteur, HIV, Inflammation and Persistence, 75015 Paris, France.
| |
Collapse
|
34
|
Wang J, Pan TZ, Huang PP, Sun ZM, Zhu HP. [Correlation between immune reconstitution and chronic graft-versus-host disease after unrelated cord blood transplantation and sibling peripheral blood stem cell transplantation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:466-473. [PMID: 34384152 PMCID: PMC8295618 DOI: 10.3760/cma.j.issn.0253-2727.2021.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Indexed: 11/05/2022]
Abstract
Objective: To explore the relationship between the reconstitution of immune cells in patients with hematological malignancies and the occurrence of chronic graft-versus-host disease (cGVHD) after treatment with unrelated cord blood transplantation (UCBT) and sibling peripheral blood stem cell transplantation (PBSCT) . Methods: A total of 124 patients undergoing allogenic hematopoietic stem cell transplantation (allo-HSCT) in the First Affiliated Hospital of University of Science and Technology of China from March 2018 to August 2019, including 96 patients with UCBT and 28 patients with PBSCT. Peripheral blood immune cells of patients with UCBT and PBSCT were detected at 1, 3, 6, 9, and 12 months after transplantation using flow cytometry, and both UCBT and PBSCT patients were divided into cGVHD and non-cGVHD groups based on whether cGVHD occurred to explore the correlation between the immune cells reconstitution of the two types of transplantation and cGVHD. Results: ①The cumulative incidence of the moderate to severe cGVHD in the UCBT group was significantly lower than that in the PBSCT group[9.38% (95% CI 3.35%-15.02%) vs 28.57% (95% CI 9.72%-43.50%) , P=0.008]; the 2-year cumulative incidence of cGVHD and moderate to severe cGVHD in the UCBT group was lower than that in the PBSCT group[15.60% (95% CI 9.20%-23.60%) vs 32.10% (95% CI 15.80%-49.70%) , P=0.047; 10.40% (95% CI 5.30%-17.50%) vs 28.60% (95% CI 13.30%-46.00%) , P=0.014]. ②The absolute counts of CD4(+)T cells in the UCBT group were higher than those in the PBSCT group at 6, 9, and 12 months after transplantation[59.00 (36.70-89.65) ×10(7)/L vs 31.40 (18.10-44.00) ×10(7)/L, P<0.001; 71.30 (49.60-101.45) ×10(7)/L vs 41.60 (25.82-56.27) ×10(7)/L, P<0.001; 83.00 (50.17-121.55) ×10(7)/L vs 44.85 (31.62-62.10) ×10(7)/L, P<0.001]; the proportions of CD4(+)T cells in the UCBT group were always higher than those in the PBSCT group (P<0.05) . The absolute counts and proportions of B cells in the PBSCT group were higher than those in the UCBT group at the first month after transplantation[0.70 (0.30-1.70) ×10(7)/L vs 0.10 (0-0.30) ×10(7)/L, P<0.001; 0.45% (0.30%-2.20%) vs 0.20% (0.10%-0.40%) , P=0.002]; the absolute counts and proportions of B cells in the UCBT group were higher than those in the PBSCT group at 9 and 12 months after transplantation[53.80 (28.00-103.20) ×10(7)/L vs 23.35 (5.07-35.00) ×10(7)/L, P<0.001; 21.45 (11.80-30.45) % vs 9.00% (3.08%-16.73%) , P<0.001. 66.70 (36.97-98.72) ×10(7)/L vs 20.85 (7.72-39.40) ×10(7)/L, P<0.001; 22.20% (14.93%-29.68%) vs 8.75% (5.80%-18.93%) , P<0.001]. The absolute counts and proportions of regulatory B (Breg) cells in the UCBT group were higher than those in the PBSCT group at 6, 9, and 12 months after transplantation[1.23 (0.38-3.52) ×10(7)/L vs 0.05 (0-0.84) ×10(7)/L, P<0.001; 5.35% (1.90%-12.20%) vs 1.45% (0-7.78%) , P=0.002. 2.25 (1.07-6.71) ×10(7)/L vs 0.12 (0-0.77) ×10(7)/L, P<0.001; 6.25% (2.00%-12.33%) vs 0.80% (0-5.25%) , P<0.001. 3.69 (0.83-8.66) ×10(7)/L vs 0.46 (0-0.93) ×10(7)/L, P<0.001; 6.15% (1.63%-11.75%) vs 1.40% (0.18%-5.85%) , P<0.001].The absolute counts and proportions of CD3(+)T cells, CD8(+)T cells, and Treg cells in the UCBT group were not significantly different from those in the PBSCT group. ③The absolute counts of B cells in the non-cGVHD group of UCBT patients were higher than those in the moderate to severe cGVHD group at 6 and 12 months after transplantation (P=0.038, P=0.043) ; the proportions of B cells in the non-cGVHD group were higher than those in the moderate to severe cGVHD group at 6 months after transplantation (P=0.049) . The absolute counts of Breg cells in the non-cGVHD group of patients with UCBT were higher than those in the moderate to severe cGVHD group at 6, 9, and 12 months after transplantation (P=0.006, P=0.028, P=0.050) ; the proportions of Breg cells in the non-cGVHD group were higher than those in the moderate to severe cGVHD group at 9 months after transplantation (P=0.038) . ④The absolute counts and proportions of B and Breg cells in the non-cGVHD group of patients with PBSCT were not statistically different than those in the moderate to severe cGVHD group. Conclusion: In the process of immune cell reconstitution, the Breg cells in the UCBT group were higher than those in the PBSCT group, and the Breg cells in the non-cGVHD group of the two types of transplantation were always higher than those in the moderate to severe cGVHD group, indicating that Breg cells can reduce the occurrence of cGVHD, revealing the possible reason for the lower incidence of cGVHD in the UCBT group.
Collapse
Affiliation(s)
- J Wang
- The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital) , Hefei 230001, China
| | - T Z Pan
- The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital) , Hefei 230001, China
| | - P P Huang
- The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital) , Hefei 230001, China
| | - Z M Sun
- The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital) , Hefei 230001, China Institute of Blood and Cell Therapy, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - H P Zhu
- The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital) , Hefei 230001, China Institute of Blood and Cell Therapy, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| |
Collapse
|
35
|
Klyuchnikov E, Badbaran A, Massoud R, Fritsche-Friedland U, Janson D, Ayuk F, Wolschke C, Bacher U, Kröger N. Enhanced Immune Reconstitution of γδ T Cells after Allogeneic Stem Cell Transplantation Overcomes the Negative Impact of Pretransplantation Minimal Residual Disease-Positive Status in Patients with Acute Myelogenous Leukemia. Transplant Cell Ther 2021; 27:841-850. [PMID: 34118468 DOI: 10.1016/j.jtct.2021.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/02/2021] [Accepted: 06/02/2021] [Indexed: 01/08/2023]
Abstract
Minimal/measurable residual disease (MRD) before allogeneic stem cell transplantation (allo-SCT) in patients with acute myelogenous leukemia (AML) is a poor risk factor for outcome. γδ T cells represent a unique minority lymphocyte population that is preferentially located in peripheral tissues, can recognize antigens in a non-MHC-restricted manner, and plays a "bridging" role between the innate and adaptive immune systems. In this study, we investigated a potential graft-versus-leukemia effect of γδ T cell reconstitution post-transplantation in AML patients with pretransplantation positive MRD status (MRD+). MRD assessment was performed in 202 patients using multicolored flow cytometry ("different from normal" strategy); 100 patients were deemed MRD+. Analysis for absolute concentrations of CD3+, CD4+, CD8+, natural killer, and γδ T cells were performed by flow cytometry according to an internal protocol at day +30 and +100 post-transplantation. Differences between categorical and continuous variables were determined using the chi-square and Student t test, respectively. The Mann-Whitney U test was used to compare medians of continuous variables. Spearman's correlation was used for nonparametric assessment of correlation between different cell subsets during immune reconstitution. Kaplan-Meier survival analysis and Cox regression analysis were used to investigate the associations between immune reconstitution and survival outcomes. Gray's analysis was used to compute incidences of relapse, nonrelapse mortality, and graft-versus-host disease (GVHD). The median follow-up of survivors was 28 months (range 3 to 59 months). Younger age (≤58 years) of recipient and donor (<30 years), sex mismatch, use of a matched donor, cytomegalovirus reactivation, and administration of antithymocyte globulin were associated with a faster γδ T cell reconstitution. In multivariable analysis for MRD+ patients, a higher than median level of γδ T cells on days +30 and +100 resulted in significantly improved leukemia-free survival (hazard ratio [HR], 0.42 [P = .007] and 0.42 [P = .011], respectively) and overall survival (HR, 0.44 [P = .038] and 0.33 [P = .009], respectively). Furthermore, a higher γδ T cell level on day +30 was associated with a significantly reduced risk of relapse (HR, 0.36; P = .019). No impact of γδ T cell level on relapse at days +30 and +100 could be seen in MRD-negative patients, and no correlation with occurrence of GVHD was observed. Our data indicate that enhanced immune reconstitution of γδ T cells post-transplantation may overcome the higher relapse risk of pretransplantation MRD+ status in patients with AML.
Collapse
Affiliation(s)
- Evgeny Klyuchnikov
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Badbaran
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Radwan Massoud
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrike Fritsche-Friedland
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dietlinde Janson
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Wolschke
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrike Bacher
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, Switzerland
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
36
|
Kielsen K, Oostenbrink LVE, von Asmuth EGJ, Jansen-Hoogendijk AM, van Ostaijen-Ten Dam MM, Ifversen M, Heilmann C, Schilham MW, van Halteren AGS, Bredius RGM, Lankester AC, Jol-van der Zijde CM, van Tol MJD, Müller K. IL-7 and IL-15 Levels Reflect the Degree of T Cell Depletion during Lymphopenia and Are Associated with an Expansion of Effector Memory T Cells after Pediatric Hematopoietic Stem Cell Transplantation. THE JOURNAL OF IMMUNOLOGY 2021; 206:2828-2838. [PMID: 34108260 DOI: 10.4049/jimmunol.2001077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/22/2021] [Indexed: 11/19/2022]
Abstract
Differentially and functionally distinct T cell subsets are involved in the development of complications after allogeneic hematopoietic stem cell transplantation (HSCT), but little is known about factors regulating their recovery after HSCT. In this study, we investigated associations between immune-regulating cytokines, T cell differentiation, and clinical outcomes. We included 80 children undergoing allogeneic HSCT for acute leukemia using bone marrow or peripheral blood stem cells grafted from a matched sibling or unrelated donor. Cytokines (IL-7, IL-15, IL-18, SCF, IL-6, IL-2, and TNF-α) and active anti-thymocyte globulin (ATG) levels were longitudinally measured along with extended T cell phenotyping. The cytokine profiles showed a temporary rise in IL-7 and IL-15 during lymphopenia, which was strongly dependent on exposure to active ATG. High levels of IL-7 and IL-15 from graft infusion to day +30 were predictive of slower T cell recovery during the first 2 mo post-HSCT; however, because of a major expansion of memory T cell stages, only naive T cells remained decreased after 3 mo (p < 0.05). No differential effect was seen on polarization of CD4+ T cells into Th1, Th2, or Th17 cells or regulatory T cells. Low levels of IL-7 and IL-15 at day +14 were associated with acute graft-versus-host disease grades II-IV in ATG-treated patients (p = 0.0004 and p = 0.0002, respectively). Children with IL-7 levels comparable to healthy controls at day +14 post-HSCT were less likely to develop EBV reactivation posttransplant. These findings suggest that quantification of IL-7 and IL-15 may be useful as biomarkers in assessing the overall T cell depletion and suggest a potential for predicting complications after HSCT.
Collapse
Affiliation(s)
- Katrine Kielsen
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; .,Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; and
| | - Lisa V E Oostenbrink
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Erik G J von Asmuth
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Anja M Jansen-Hoogendijk
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Monique M van Ostaijen-Ten Dam
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Marianne Ifversen
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Carsten Heilmann
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Marco W Schilham
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Astrid G S van Halteren
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Robbert G M Bredius
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Arjan C Lankester
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Cornelia M Jol-van der Zijde
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Maarten J D van Tol
- Laboratory of Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Klaus Müller
- Hematopoietic Stem Cell Transplantation and Primary Immune Deficiency, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Disease, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; and
| |
Collapse
|
37
|
Yeo GEC, Ng MH, Nordin FB, Law JX. Potential of Mesenchymal Stem Cells in the Rejuvenation of the Aging Immune System. Int J Mol Sci 2021; 22:5749. [PMID: 34072224 PMCID: PMC8198707 DOI: 10.3390/ijms22115749] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Rapid growth of the geriatric population has been made possible with advancements in pharmaceutical and health sciences. Hence, age-associated diseases are becoming more common. Aging encompasses deterioration of the immune system, known as immunosenescence. Dysregulation of the immune cell production, differentiation, and functioning lead to a chronic subclinical inflammatory state termed inflammaging. The hallmarks of the aging immune system are decreased naïve cells, increased memory cells, and increased serum levels of pro-inflammatory cytokines. Mesenchymal stem cell (MSC) transplantation is a promising solution to halt immunosenescence as the cells have excellent immunomodulatory functions and low immunogenicity. This review compiles the present knowledge of the causes and changes of the aging immune system and the potential of MSC transplantation as a regenerative therapy for immunosenescence.
Collapse
Affiliation(s)
| | | | | | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Malaysia; (G.E.C.Y.); (M.H.N.); (F.B.N.)
| |
Collapse
|
38
|
Lin RJ, Elias HK, van den Brink MRM. Immune Reconstitution in the Aging Host: Opportunities for Mechanism-Based Therapy in Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2021; 12:674093. [PMID: 33953731 PMCID: PMC8089387 DOI: 10.3389/fimmu.2021.674093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
Older patients with hematologic malignancies are increasingly considered for allogeneic hematopoietic cell transplantation with encouraging outcomes. While aging-related thymic dysfunction remains a major obstacle to optimal and timely immune reconstitution post- transplantation, recent accumulating evidence has suggested that various aging hallmarks such as cellular senescence, inflamm-aging, and hematopoietic stem cell exhaustion, could also impact immune reconstitution post-transplantation in both thymic-dependent and independent manner. Here we review molecular and cellular aspects of immune senescence and immune rejuvenation related to allogeneic hematopoietic cell transplantation among older patients and discuss potential strategies for mechanism-based therapeutic intervention.
Collapse
Affiliation(s)
- Richard J Lin
- Adult Bone Marrow Transplantation (BMT) Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Harold K Elias
- Adult Bone Marrow Transplantation (BMT) Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation (BMT) Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
39
|
Granadier D, Iovino L, Kinsella S, Dudakov JA. Dynamics of thymus function and T cell receptor repertoire breadth in health and disease. Semin Immunopathol 2021; 43:119-134. [PMID: 33608819 PMCID: PMC7894242 DOI: 10.1007/s00281-021-00840-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/12/2021] [Indexed: 12/26/2022]
Abstract
T cell recognition of unknown antigens relies on the tremendous diversity of the T cell receptor (TCR) repertoire; generation of which can only occur in the thymus. TCR repertoire breadth is thus critical for not only coordinating the adaptive response against pathogens but also for mounting a response against malignancies. However, thymic function is exquisitely sensitive to negative stimuli, which can come in the form of acute insult, such as that caused by stress, infection, or common cancer therapies; or chronic damage such as the progressive decline in thymic function with age. Whether it be prolonged T cell deficiency after hematopoietic cell transplantation (HCT) or constriction in the breadth of the peripheral TCR repertoire with age; these insults result in poor adaptive immune responses. In this review, we will discuss the importance of thymic function for generation of the TCR repertoire and how acute and chronic thymic damage influences immune health. We will also discuss methods that are used to measure thymic function in patients and strategies that have been developed to boost thymic function.
Collapse
Affiliation(s)
- David Granadier
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Department of Molecular and Cellular Biology, University of Washington, Seattle, WA, USA
| | - Lorenzo Iovino
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sinéad Kinsella
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jarrod A Dudakov
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
40
|
Huo YY, Pang AM, Cheng T. [Advance in hematopoietic and immune reconstitution of allogeneic stem cell transplantation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 41:958-963. [PMID: 33333706 PMCID: PMC7767801 DOI: 10.3760/cma.j.issn.0253-2727.2020.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Y Y Huo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - A M Pang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - T Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
41
|
Irla M. RANK Signaling in the Differentiation and Regeneration of Thymic Epithelial Cells. Front Immunol 2021; 11:623265. [PMID: 33552088 PMCID: PMC7862717 DOI: 10.3389/fimmu.2020.623265] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/07/2020] [Indexed: 11/24/2022] Open
Abstract
Thymic epithelial cells (TECs) provide essential clues for the proliferation, survival, migration, and differentiation of thymocytes. Recent advances in mouse and human have revealed that TECs constitute a highly heterogeneous cell population with distinct functional properties. Importantly, TECs are sensitive to thymic damages engendered by myeloablative conditioning regimen used for bone marrow transplantation. These detrimental effects on TECs delay de novo T-cell production, which can increase the risk of morbidity and mortality in many patients. Alike that TECs guide the development of thymocytes, reciprocally thymocytes control the differentiation and organization of TECs. These bidirectional interactions are referred to as thymic crosstalk. The tumor necrosis factor receptor superfamily (TNFRSF) member, receptor activator of nuclear factor kappa-B (RANK) and its cognate ligand RANKL have emerged as key players of the crosstalk between TECs and thymocytes. RANKL, mainly provided by positively selected CD4+ thymocytes and a subset of group 3 innate lymphoid cells, controls mTEC proliferation/differentiation and TEC regeneration. In this review, I discuss recent advances that have unraveled the high heterogeneity of TECs and the implication of the RANK-RANKL signaling axis in TEC differentiation and regeneration. Targeting this cell-signaling pathway opens novel therapeutic perspectives to recover TEC function and T-cell production.
Collapse
Affiliation(s)
- Magali Irla
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| |
Collapse
|
42
|
Tickotsky-Moskovitz N, Louzoun Y, Dvorkin S, Rotkopf A, Kuperman AA, Efroni S. CDR3 and V genes show distinct reconstitution patterns in T cell repertoire post-allogeneic bone marrow transplantation. Immunogenetics 2021; 73:163-173. [PMID: 33475766 DOI: 10.1007/s00251-020-01200-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
Restoration of T cell repertoire diversity after allogeneic bone marrow transplantation (allo-BMT) is crucial for immune recovery. T cell diversity is produced by rearrangements of germline gene segments (V (D) and J) of the T cell receptor (TCR) α and β chains, and selection induced by binding of TCRs to MHC-peptide complexes. Multiple measures were proposed for this diversity. We here focus on the V-gene usage and the CDR3 sequences of the beta chain. We compared multiple T cell repertoires to follow T cell repertoire changes post-allo-BMT in HLA-matched related donor and recipient pairs. Our analyses of the differences between donor and recipient complementarity determining region 3 (CDR3) beta composition and V-gene profile show that the CDR3 sequence composition does not change during restoration, implying its dependence on the HLA typing. In contrast, V-gene usage followed a time-dependent pattern, initially following the donor profile and then shifting back to the recipients' profile. The final long-term repertoire was more similar to that of the recipient's original one than the donor's; some recipients converged within months, while others took multiple years. Based on the results of our analyses, we propose that donor-recipient V-gene distribution differences may serve as clinical biomarkers for monitoring immune recovery.
Collapse
Affiliation(s)
| | - Yoram Louzoun
- Department of Mathematics, Bar Ilan University, Ramat Gan, Israel.
| | - Shirit Dvorkin
- Department of Mathematics, Bar Ilan University, Ramat Gan, Israel
| | - Adi Rotkopf
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Amir Asher Kuperman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Blood Coagulation Service and Pediatric Hematology Clinic, Galilee Medical Center, Nahariya, Israel
| | - Sol Efroni
- The Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
| |
Collapse
|
43
|
Wölfl M, Qayed M, Benitez Carabante MI, Sykora T, Bonig H, Lawitschka A, Diaz-de-Heredia C. Current Prophylaxis and Treatment Approaches for Acute Graft-Versus-Host Disease in Haematopoietic Stem Cell Transplantation for Children With Acute Lymphoblastic Leukaemia. Front Pediatr 2021; 9:784377. [PMID: 35071133 PMCID: PMC8771910 DOI: 10.3389/fped.2021.784377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Acute graft-versus-host disease (aGvHD) continues to be a leading cause of morbidity and mortality following allogeneic haematopoietic stem cell transplantation (HSCT). However, higher event-free survival (EFS) was observed in patients with acute lymphoblastic leukaemia (ALL) and grade II aGvHD vs. patients with no or grade I GvHD in the randomised, controlled, open-label, international, multicentre Phase III For Omitting Radiation Under Majority age (FORUM) trial. This finding suggests that moderate-severity aGvHD is associated with a graft-versus-leukaemia effect which protects against leukaemia recurrence. In order to optimise the benefits of HSCT for leukaemia patients, reduction of non-relapse mortality-which is predominantly caused by severe GvHD-is of utmost importance. Herein, we review contemporary prophylaxis and treatment options for aGvHD in children with ALL and the key challenges of aGvHD management, focusing on maintaining the graft-versus-leukaemia effect without increasing the severity of GvHD.
Collapse
Affiliation(s)
- Matthias Wölfl
- Pediatric Hematology, Oncology and Stem Cell Transplantation, Children's Hospital, Würzburg University Hospital, Würzburg, Germany
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, United States
| | - Maria Isabel Benitez Carabante
- Department of Pediatric Hematology and Oncology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Tomas Sykora
- Haematopoietic Stem Cell Transplantation Unit, Department of Pediatric Haematology and Oncology, Comenius University Children's Hospital, Bratislava, Slovakia
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe-University Frankfurt/Main, Frankfurt, Germany.,German Red Cross Blood Service BaWüHe, Frankfurt, Germany
| | - Anita Lawitschka
- Department of Pediatrics, St. Anna Kinderspital and Children's Cancer Research Institute, Medical University of Vienna, Vienna, Austria
| | - Cristina Diaz-de-Heredia
- Department of Pediatric Hematology and Oncology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| |
Collapse
|
44
|
Camacho-Bydume C, Wang T, Sees JA, Fernandez-Viña M, Abid MB, Askar M, Beitinjaneh A, Brown V, Castillo P, Chhabra S, Gadalla SM, Hsu JM, Kamoun M, Lazaryan A, Nishihori T, Page K, Schetelig J, Fleischhauer K, Marsh SGE, Paczesny S, Spellman SR, Lee SJ, Hsu KC. Specific Class I HLA Supertypes but Not HLA Zygosity or Expression Are Associated with Outcomes following HLA-Matched Allogeneic Hematopoietic Cell Transplant: HLA Supertypes Impact Allogeneic HCT Outcomes. Transplant Cell Ther 2020; 27:142.e1-142.e11. [PMID: 33053450 DOI: 10.1016/j.bbmt.2020.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Maximizing the probability of antigen presentation to T cells through diversity in HLAs can enhance immune responsiveness and translate into improved clinical outcomes, as evidenced by the association of heterozygosity and supertypes at HLA class I loci with improved survival in patients with advanced solid tumors treated with immune checkpoint inhibitors. We investigated the impact of HLA heterozygosity, supertypes, and surface expression on outcomes in adult and pediatric patients with acute myeloid leukemia (AML), myelodysplastic syndrome, acute lymphoblastic leukemia, and non-Hodgkin lymphoma who underwent 8/8 HLA-matched, T cell replete, unrelated, allogeneic hematopoietic cell transplant (HCT) from 2000 to 2015 using patient data reported to the Center for International Blood and Marrow Transplant Research. HLA class I heterozygosity and HLA expression were not associated with overall survival, relapse, transplant-related mortality (TRM), disease-free survival (DFS), and acute graft-versus-host disease following HCT. The HLA-B62 supertype was associated with decreased TRM in the entire patient cohort (hazard ratio [HR], 0.79; 95% CI, 0.69 to 0.90; P = .00053). The HLA-B27 supertype was associated with worse DFS in patients with AML (HR = 1.21; 95% CI, 1.10 to 1.32; P = .00005). These findings suggest that the survival benefit of HLA heterozygosity seen in solid tumor patients receiving immune checkpoint inhibitors does not extend to patients undergoing allogeneic HCT. Certain HLA supertypes, however, are associated with TRM and DFS, suggesting that similarities in peptide presentation between supertype members play a role in these outcomes. Beyond implications for prognosis following HCT, these findings support the further investigation of these HLA supertypes and the specific immune peptides important for transplant outcomes.
Collapse
Affiliation(s)
| | - Tao Wang
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI
| | - Jennifer A Sees
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be The Match, Minneapolis, MN
| | | | - Muhammad Bilal Abid
- Divisions of Hematology/Oncology and Infectious Diseases, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Medhat Askar
- Department of Pathology and Laboratory Medicine, Baylor University Medical Center, Dallas, Texas
| | - Amer Beitinjaneh
- Department of Medicine, Division of Transplantation and Cellular Therapy, University of Miami, Miami, Florida
| | - Valerie Brown
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Penn State Hershey Children's Hospital and College of Medicine, Hershey, Pennsylvania
| | - Paul Castillo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Florida Health Shands Children's Hospital, Gainesville, FL
| | - Saurabh Chhabra
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Shahinaz M Gadalla
- Division of Cancer Epidemiology & Genetics, NIH-NCI Clinical Genetics Branch, Rockville, Maryland
| | - Jing-Mei Hsu
- Division of Hematology/Oncology, Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine/New York Presbyterian Hospital, New York, NY
| | - Malek Kamoun
- Deparment of Pathology and Laboratory Medicine, Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Aleksandr Lazaryan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, Florida
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, Tampa, Florida
| | - Kristin Page
- Division of Pediatric Blood and Marrow Transplantation, Duke University Medical Center, Durham, North Carolina
| | - Johannes Schetelig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | | | - Steven G E Marsh
- Anthony Nolan Research Institute, Royal Free Hospital, London, UK; UCL Cancer Institute, London, UK
| | - Sophie Paczesny
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Stephen R Spellman
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be The Match, Minneapolis, MN
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, WA
| | - Katharine C Hsu
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York; Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
45
|
Bhatt ST, Bednarski JJ. Immune Reconstitution in Pediatric Patients Following Hematopoietic Cell Transplant for Non-malignant Disorders. Front Immunol 2020; 11:1988. [PMID: 33013851 PMCID: PMC7461808 DOI: 10.3389/fimmu.2020.01988] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/23/2020] [Indexed: 01/24/2023] Open
Abstract
Allogeneic hematopoietic cell transplant (HCT) is curative for pediatric patients with non-malignant hematopoietic disorders, including hemoglobinopathies, bone marrow failure syndromes, and primary immunodeficiencies. Early establishment of donor-derived innate and adaptive immunity following HCT is associated with improved overall survival, lower risk of infections and decreased incidence of graft failure. Immune reconstitution (IR) is impacted by numerous clinical variables including primary disease, donor characteristics, conditioning regimen, and graft versus host disease (GVHD). Recent advancements in HCT have been directed at reducing toxicity of conditioning therapy, expanding donor availability through use of alternative donor sources, and addressing morbidity from GVHD with novel graft manipulation. These novel transplant approaches impact the kinetics of immune recovery, which influence post-transplant outcomes. Here we review immune reconstitution in pediatric patients undergoing HCT for non-malignant disorders. We explore the transplant-associated factors that influence immunologic recovery and the disease-specific associations between IR and transplant outcomes.
Collapse
Affiliation(s)
- Sima T Bhatt
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| |
Collapse
|
46
|
Dumont-Lagacé M, Li Q, Tanguay M, Chagraoui J, Kientega T, Cardin GB, Brasey A, Trofimov A, Carli C, Ahmad I, Bambace NM, Bernard L, Kiss TL, Roy J, Roy DC, Lemieux S, Perreault C, Rodier F, Dufresne SF, Busque L, Lachance S, Sauvageau G, Cohen S, Delisle JS. UM171-Expanded Cord Blood Transplants Support Robust T Cell Reconstitution with Low Rates of Severe Infections. Transplant Cell Ther 2020; 27:76.e1-76.e9. [PMID: 33022376 DOI: 10.1016/j.bbmt.2020.09.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 10/23/2022]
Abstract
Rapid T cell reconstitution following hematopoietic stem cell transplantation (HSCT) is essential for protection against infections and has been associated with lower incidence of chronic graft-versus-host disease (cGVHD), relapse, and transplant-related mortality (TRM). While cord blood (CB) transplants are associated with lower rates of cGVHD and relapse, their low stem cell content results in slower immune reconstitution and higher risk of graft failure, severe infections, and TRM. Recently, results of a phase I/II trial revealed that single UM171-expanded CB transplant allowed the use of smaller CB units without compromising engraftment (www.clinicaltrials.gov, NCT02668315). We assessed T cell reconstitution in patients who underwent transplantation with UM171-expanded CB grafts and retrospectively compared it to that of patients receiving unmanipulated CB transplants. While median T cell dose infused was at least 2 to 3 times lower than that of unmanipulated CB, numbers and phenotype of T cells at 3, 6, and 12 months post-transplant were similar between the 2 cohorts. T cell receptor sequencing analyses revealed that UM171 patients had greater T cell diversity and higher numbers of clonotypes at 12 months post-transplant. This was associated with higher counts of naive T cells and recent thymic emigrants, suggesting active thymopoiesis and correlating with the demonstration that UM171 expands common lymphoid progenitors in vitro. UM171 patients also showed rapid virus-specific T cell reactivity and significantly reduced incidence of severe infections. These results suggest that UM171 patients benefit from rapid T cell reconstitution, which likely contributes to the absence of moderate/severe cGVHD, infection-related mortality, and late TRM observed in this cohort.
Collapse
Affiliation(s)
- Maude Dumont-Lagacé
- ExCellThera, Inc., Montreal, Quebec, Canada; Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada
| | - Qi Li
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Mégane Tanguay
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada
| | - Jalila Chagraoui
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada
| | - Tibila Kientega
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Guillaume B Cardin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Ann Brasey
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Assya Trofimov
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada; Department of Computer Science and Operations Research, Université de Montréal, Montreal, Quebec, Canada
| | - Cédric Carli
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Imran Ahmad
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Nadia M Bambace
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Léa Bernard
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Thomas L Kiss
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Jean Roy
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Denis-Claude Roy
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Sébastien Lemieux
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada; Department of Computer Science and Operations Research, Université de Montréal, Montreal, Quebec, Canada.; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montreal, Quebec, Canada; Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Simon Frédéric Dufresne
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, Quebec, Canada; Division of Infectious Diseases and Clinical Microbiology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Lambert Busque
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Silvy Lachance
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Guy Sauvageau
- ExCellThera, Inc., Montreal, Quebec, Canada; Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Sandra Cohen
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Jean-Sébastien Delisle
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada.
| |
Collapse
|
47
|
PTCy ameliorates GVHD by restoring regulatory and effector T-cell homeostasis in recipients with PD-1 blockade. Blood Adv 2020; 3:4081-4094. [PMID: 31821459 DOI: 10.1182/bloodadvances.2019000134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
Graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a significant cause of morbidity and mortality. Regulatory T cells (Tregs) are critical mediators of immune tolerance after allo-HSCT. Clinical studies have indicated that programmed cell death 1 (PD-1) blockade before allo-HSCT involves a risk of severe GVHD. However, the mechanisms underlying GVHD induction resulting from PD-1 blockade remain unclear. We investigated the impact of PD-1 expression of donor T cells on T-cell reconstitution and GVHD using murine models. We first demonstrated that inhibition of PD-1 signaling induced aggressive expansion of CD4+ conventional T cells; however, Tregs could not maintain expansion because of high susceptibility to apoptosis, resulting in discordant immune recovery and subsequent development of severe GVHD. We then evaluated the impact of posttransplantation cyclophosphamide (PTCy) on abnormal T-cell reconstitution after PD-1 blockade. PTCy efficiently ameliorated GVHD after transplantation from a PD-1-/- donor and extended overall survival by safely regulating the proliferation and apoptosis of T-cell subsets. Notably, in the first 2 weeks after administration of PTCy, Tregs regained their ability to continuously proliferate, resulting in well-balanced reconstitution of donor T-cell subsets. In conclusion, the influence of PD-1 blockade differed within T-cell subsets and caused unbalanced reconstitution of T-cell subsets, resulting in severe GVHD. PTCy successfully restored T-cell homeostasis and ameliorated GVHD induced by PD-1-/- donor T cells. These findings may help explain the pathophysiology behind the observation that PTCy may mitigate the incidence and impact of GVHD associated with prior exposure to PD-1 blockade.
Collapse
|
48
|
Haematopoietic stem cell transplantation in systemic sclerosis: Challenges and perspectives. Autoimmun Rev 2020; 19:102662. [PMID: 32942028 DOI: 10.1016/j.autrev.2020.102662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Systemic Sclerosis is chronic progressive autoimmune disease, characterised by microangiopathy and fibrosis. Due to disease heterogeneity, in terms of extent, severity, and rate of progression, optimal therapeutic interventions are still lacking. Haematopoietic stem cells may be a new therapeutic option in this disease and, although the results of the first trials are encouraging, several issues remain to be addressed. On these bases, the stem cells transplantation is an area of active investigation, and an overview of the current available literature may help to define the role of this therapeutic strategy. Although the promising results, some unmet needs remain, including the transplantation protocols and their effects on immune system, the selection of the ideal patient and the pre-transplant cardiopulmonary evaluations. An improvement in these fields will allow us to optimize the haematopoietic stem cell therapies in SSc.
Collapse
|
49
|
André I, Simons L, Ma K, Moirangthem RD, Diana JS, Magrin E, Couzin C, Magnani A, Cavazzana M. Ex vivo generated human T-lymphoid progenitors as a tool to accelerate immune reconstitution after partially HLA compatible hematopoietic stem cell transplantation or after gene therapy. Bone Marrow Transplant 2020; 54:749-755. [PMID: 31431705 DOI: 10.1038/s41409-019-0599-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prolonged T-cell immunodeficiency following HLA- incompatible hematopoietic stem cell transplantation (HSCT) represents a major obstacle hampering the more widespread use of this approach. Strategies to fasten T-cell reconstitution in this setting are highly warranted as opportunistic infections and an increased risk of relapse account for high rates of morbidity and mortality especially during early month following this type of HSCT. We have implemented a feeder free cell system based on the use of the notch ligand DL4 and cytokines allowing for the in vitro differentiation of human T-Lymphoid Progenitor cells (HTLPs) from various sources of CD34+ hematopoietic stem and precursor cells (HSPCs). Co- transplantion of human T-lymphoid progenitors (HTLPs) and non- manipulated HSPCs into immunodeficient mice successfully accelerated the reconstitution of a polyclonal T-cell repertoire. This review summarizes preclinical data on the use of T-cell progenitors for treatment of post- transplantation immunodeficiency and gives insights into the development of GMP based protocols for potential clinical applications including gene therapy approaches. Future clinical trials implementing this protocol will aim at the acceleration of immune reconstitution in different clinical settings such as SCID and leukemia patients undergoing allogeneic transplantation. Apart from pure cell-therapy approaches, the combination of DL-4 culture with gene transduction protocols will open new perspectives in terms of gene therapy applications for primary immunodeficiencies.
Collapse
Affiliation(s)
- Isabelle André
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France. .,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France. .,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France.
| | - Laura Simons
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Kuiying Ma
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Ranjita Devi Moirangthem
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Jean-Sébastien Diana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Elisa Magrin
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Chloé Couzin
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alessandra Magnani
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marina Cavazzana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University - Sorbonne Paris Cité, Imagine Institute, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
50
|
Legoff J, Michonneau D, Socie G. The virome in hematology-Stem cell transplantation and beyond. Semin Hematol 2020; 57:19-25. [PMID: 32690140 DOI: 10.1053/j.seminhematol.2020.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 05/06/2020] [Indexed: 12/23/2022]
Abstract
The virome has been recently studied in hematology and mostly in the setting of allogeneic hematopoietic stem cell transplantation. However, in hematology (as in the setting of nonhematological disorders) the study of the microbiome (that indeed includes the virome) is a growing field. The overall field is moving beyond species catalogue to the understanding of the complex ecological relationship that microbes have with each other and with their host. Here we review the existing literature on the virome in transplant recipients and in other settings, and discuss potential applications of the virome study in hematology.
Collapse
Affiliation(s)
- Jérôme Legoff
- Université de Paris, INSERM U976, Paris, France; Microbiology department, Virology laboratory, Saint Louis Hospital, Paris, France
| | - David Michonneau
- Université de Paris, INSERM U976, Paris, France; Hematology-Transplantation, Saint Louis Hospital, Paris, France
| | - Gérard Socie
- Université de Paris, INSERM U976, Paris, France; Hematology-Transplantation, Saint Louis Hospital, Paris, France.
| |
Collapse
|